Bone Marrow Biopsy Pathology: Christine Beham-Schmid Annette Schmitt-Graeff
Bone Marrow Biopsy Pathology: Christine Beham-Schmid Annette Schmitt-Graeff
Christine Beham-Schmid
Annette Schmitt-Graeff
Series Editor
Farid Moinfar
Department of Pathology, Ordensklinikum Linz/Hospital of the Sisters of Charity
Department of Pathology, Medical University of Graz
Graz, Austria
This Springer imprint is published by the registered company Springer-Verlag GmbH, DE part of Springer Nature.
The registered company address is: Heidelberger Platz 3, 14197 Berlin, Germany
Preface
When I (C.B-S) was asked by Prof. F. Moinfar to write a book on bone marrow pathology, I
was hesitant in view of the abundance of excellent textbooks on this topic. However, the fact
that this book should be intended as a practical guide dominated rather by illustrations than by
text, and the promise of Prof. Annette Schmitt-Graeff to act as a coauthor, at least encouraged
me to start this project.
    As a young internship doctor in the small public hospital in Oberndorf/Salzburg, I (C.B-S)
was stimulated by Primarius Dr. K. Mittermayer to have a look on bone marrow aspirates. This
resulted in my interest in hematology, which was deepened when I consecutively started a
training in pathology with focus on hematopathology at the Institute of Pathology, Faculty of
Medicine, University of Graz. During this time, I received intensive training in bone marrow
biopsy pathology in Munich (Prof. Burkhardt) and Tel Aviv (Bertha Frisch) and in lymphoma
diagnostics in London (P.G. Isaacson). Up to now, I have seen ten thousands of bone marrow
biopsies and gained a lot of experience. Since the knowledge of clinical data is essential for
exact pathohistological diagnosis of bone marrow biopsies, a close cooperation with the clini-
cal colleagues is mandatory.
    My (A.S-G) enthusiasm for bone marrow (BM) diagnostics dates back to my residency in
hematology/oncology at the Departments of Internal Medicine, University Hospitals of
Cologne and Essen, Germany. I was keen on evaluating blood and BM cytology. I also realized
the impact of BM and lymph node biopsies on therapeutic strategies for our patients suffering
from myeloid or lymphoproliferative neoplasms. This appreciation was the rationale to pursue
a residency and a fellowship in anatomical pathology at the University Hospital Düsseldorf,
Germany, where I established histologic BM assessment. My research activities were on the
cytoskeleton including BM stroma with Prof. Dr. Dr. h.c. Giulio Gabbiani, University Hospital
Geneva, Switzerland, where I assumed the position as consultant. There and at the Berlin
Reference Center for Lymph Node Pathology of Prof. Dr. Dr. h.c. Harald Stein I was princi-
pally in charge of BM pathology including supervising residents in this field.
    Later, I joined the faculty at the Medical Center, University Freiburg, Germany, where I was
appointed Professor of Hematopathology and GI Pathology. My activities were focused on
integrating morphological, immunophenotypic, and molecular aspects of hematological neo-
plasms and primary immunodeficiency disorders with the late Prof. Dr. Paul Fisch in close
clinico-pathologic cooperation.
    As previous president of the German Division of the IAP (GDIAP) and actual member of
the General IAP and GDIAP Education committees, I have taught and directed numerous
hematopathology courses of the IAP nationally and internationally. In the context of my active
involvement in education and teaching, I am delighted to contribute to this textbook with a mix
of basic information and challenging diagnostic cases.
    This book is intended as a practical guide in the complex field of bone marrow pathology.
We hope it will turn out helpful in the daily diagnostic work.
June 2020
Graz, Austria                                                       Christine Beham-Schmid
Freiburg, Germany                                                    Annette Schmitt-Graeff
                                                                                              v
Acknowledgments
I would like to thank my coauthor and friend Annette Schmitt-Graeff, without her support, my
own contributions for this book would not have been possible.
    Moreover, I am deeply indebted to Prof. Farid Moinfar for giving me the opportunity to
write this book with my coauthor.
    My special acknowledgment is also attributed to my teachers in hematology and hematopa-
thology Prim. Dr. K. Mittermayer, Oberndorf/Salzburg, Prof. Dr. R. Burkhardt, Munich/
Germany, Prof. Dr. B. Frisch, Tel Aviv/Israel, Prof. P.G. Isaacson, London/G.B., and Prof. Dr.
J. Thiele, Cologne/Germany.
    In addition, I would like to recognize the excellent work of the team of the laboratory of
hematopathology and the photographers, Institute of Pathology, Medical University of Graz,
Austria.
    Last but not least, I am most grateful to my husband Alfred and my son Clemens for their
never-ending patience and support during this project.
Christine Beham-Schmid
First, I would like to acknowledge the great dedication of my coauthor Prof. Dr. Christine
Beham-Schmid, Graz, and the series Editor Prof. Dr. Farid Moinfar, Linz, Austria, to the proj-
ect of our book.
   I express my deep appreciation to many experts who inspired me with their devotion to
hematopathology and hematology. In particular, I want to mention Prof. Dr. Dr.hc Harald
Stein, Berlin, and Prof. Dr. Juergen Thiele, Cologne/Hannover, Germany, and Prof. Dr. Kristin
Henry, London, G.B. Special thanks go to Prof. Dr. Rüdiger Hehlmann who founded the
European LeukemiaNet (ELN). I would also like to acknowledge Prof. Dr. Attilio Orazi, El
Paso, and Prof. Dr. Daniel A. Arber, Chicago, USA, for their exciting investigations and teach-
ing in hematopathology.
   I am especially indebted to Prof. Dr. Robert Zeiser, Dr. Dietmar Pfeifer, Dr. Milena Pantic,
and Dr. Justina Rawluk, Department of Hematology and Oncology, Medical Center, University
Freiburg, Germany, for their support and helpful contribution to molecular analyses, cytoge-
netics, and cytology of myeloid neoplasms.
   Lastly, I would like to thank my mother Margret Gräff, my husband Eberhard Schmitt, our
son Rafael J. P. Schmitt, and my daughter-in-law Zelda Othenin for their patience and love.
Annette Schmitt-Graeff
                                                                                            vii
Contents
                                                                                                                                   ix
x                                                                                                                                                Contents
diagnosis, for example, recognition of normally present                  1.1.1     Bone Components (Fig. 1.1)
subcortical hypoplasia or non-representative tangentially
retrieved BMB. It is also important to recognize and                     Representative bone marrow biopsies show cortical bone
report changes in hematopoietic cells due to inadequate                  (cortex, compact bone) and trabecular bone (trabeculae, can-
fixation, decalcification, or staining procedures.                       cellous bone, ossicles) (Fig. 1.1a, b) with appendant bone
a b
c d
e f
Fig. 1.1 (a, b) Low magnification of representative BMBs with corti-     female with rarefication of trabecular bone (Gomori’s stain). (f) A
cal bone (left side) and trabecular bone (left biopsy: Gomori’s stain,   48-year-old female showing rarefication of trabecular bone (Gomori’s
right biopsy: Ladewig). (c) Higher magnification of (b) showing tra-     stain). (g) Severely thickened cancellous bone of a 60-year-old male
becular bone constituting the honeycomb of bone. (d) Normal bone         (Gomori’s stain). (h) BMB of a 73-year-old male with marked osteo-
structure (MGG) of a 50-year-old female. (e) BMB of a 52-year-old        sclerosis (Gomori’s stain)
1       Normal Bone Marrow                                                                                                     3
g h
cells, which are flat endothelial cells, lining trabecular and    The cytoplasmic processes of osteocytes connect with the
subcortical surface, osteoblasts, osteoclasts, and osteocytes.    processes of other osteocytes and also with osteoblasts on
   The cortex is a solid layer of compact bone of various         the surface of the bone (Fig. 1.2d–f). Thus, a circulatory net-
thickness, on the outside periosteum, on the inside the endos-    work within the osseous system, within the bone and to its
teum, a single layer of cells, are attached. The cortex mainly    surface, is built, providing the subsistence of the osteocytes
consists of lamellar bone, but some woven bone is also pres-      and enables the transfer between bone, bloodstream, and
ent. There is constant remodeling of the bone. In adults,         interstitial fluid.
remodeling of the bone mainly takes place in the subcortical         Osteoclasts are the bone-resorbing cells on or near the
regions. A new layer of bone is added by osteoblasts, while       surface of the bone (Fig. 1.2g), often interposed between
the osteoclasts resorb other areas of the bone. Up to 20–25%      endothelial and endosteal cells and also between subcortical
of the bone surface may be covered by osteoid.                    bone surface and endothelium on the trabecular surface.
   Trabecular bone constitutes the honeycomb of bone              Osteoclasts, with their villous extensions bind to matrix
(Fig. 1.1c, d) and is enclosed by the cortical bone. A rarefi-    adhesion proteins and produce resorption pits, shallow con-
cation of trabeculae (osteopenia) results in enlargement of       cavities, called Howship lacunae (Fig. 1.2h inset). The
marrow cavities (Fig. 1.1e, f). A thickening of cancellous        appearance of osteoclasts is either uni- or multinuclear.
bone, osteosclerosis, causes a decreased size of marrow           Multinuclear osteoclasts may even become larger than mega-
spaces (Fig. 1.1g, h). Semi-automatic and/or computerized         karyocytes (Fig. 1.2h). The relatively flat uni-nucleated
quantification systems for bone and bone marrow struc-            osteoclasts equally do resorb bone as do the multinucleated
tures are not generally required for the diagnosis and inter-     forms. Bone marrow biopsies of children and young adults
pretation of bone marrow biopsy sections. For routine             normally show marked bone remodeling with the presence
diagnosis, subjective assessment by naked eye of the com-         of many osteoclasts, whereas in adults and older people the
ponents of the biopsy sections in a number of fields in the       presence of many osteoclasts indicates a metabolic or a neo-
light microscope, or, if requested by the clinicians, assess-     plastic disease.
ment by use of a graticule in the ocular is recommended [3]          Mean values from healthy adults for osteoblastic index
(Fig. 1.2).                                                       (percentage of trabecular surface covered by cuboidal osteo-
   Osteoblasts with a diameter from 20 to 50 μm produce the       blasts) is 5%. The osteoclastic index (number of osteo-
bone matrix, osteoid, which gets subsequently ossified [4].       clasts/100 mm trabecular circumference) is 3–4/100 mm [4].
Osteoblasts line the surface of the cancellous bone and if        Although osteoclasts and osteoblasts share the surface of the
these cells are activated, they become cuboidal (Fig. 1.2a, b).   trabeculae (Fig. 1.2g), these cells originate from different
After mineralization of bone, some osteoblasts are trapped        stem cells, osteoclasts from hematopoietic, and osteoblasts
within the bone and transformed into osteocytes (Fig. 1.2c).      from mesenchymal stem cells.
4                                                                                                               1   Normal Bone Marrow
a b
c d
e f
Fig. 1.2 (a) Osteoblasts lining the surface of bone (MGG). (b)          of osteocyte (MGG). (g) Multinucleated osteoclast on bone surface
Cuboidal activated osteoblasts (MGG). (c) Trabecular bone with osteo-   (MGG). (h) Huge multinucleated osteoclasts. Inset reveals Howship
cyte (MGG). (d–f) Cytoplasmic processes of osteocytes connecting        lacunae (MGG)
with processes of adjacent osteocytes. Inset shows high magnification
1       Normal Bone Marrow       5
g h
1.2      Cellularity of the Marrow                               sue, or, similar to an aspirate, to a marrow cavity excluding
                                                                  bone tissue [2]. The latter procedure is comparable to mea-
While BM aspirates (BMA) and BM trephine biopsies                 surements of the cellularity of aspirated fragments. Slight
(BMTB) are excellent for assessment of lineage percentages        differences in marrow cellularity have been described in dif-
and morphology, only BMTB can provide accurate informa-           ferent sites like sternum, lumbal vertebrae, and iliac crest [4].
tion about the overall degree of cellularity and organization         The bone marrow of newborns mainly consists of cells
of all hematopoietic lineages. Also, if lymphocytic cells or      with a negligent amount of fat cells (0–<5%). With increasing
plasma cells are increased, only BMTB will reveal the pat-        age, cellularity decreases with an accelerated rate of reduc-
tern of their distribution.                                       tion beyond the age of 70. The decrease in hematopoietic
    However, it is important that not only the width of the       tissue with advanced age is due to a loss of bone substance
BMTB is adequate but also the length which ideally should         and also caused by a true loss of the amount of hematopoietic
measure at least 2 cm. Length is particularly important not       tissue. The marrow cavities consequently are filled with fat
only for the “staging” of neoplasms as to whether there is        cells. At the age of 20, the average cellularity is 70–80%, at
BM involvement but also in many infectious disease affect-        the age of 50 cellularity is 50–60%, and at 70 years and
ing BM (Fig. 1.3).                                                beyond, cellularity is reduced to 20–30% [2, 6–8] (Fig. 1.3a).
    The term “cellularity” refers to hematopoietic as well as         The spatial arrangement of hematopoiesis is a potential
fat cells and indicates the relative amount of these compo-       pitfall with regard to the normally present subcortical
nents. Normal values are age-dependent (Fig. 1.3a) with           hypoplasia (subcortical fatty tissue) (Fig. 1.3b–d). To
individual deviations [5, 6]. For routine diagnosis, the cellu-   avoid the error of estimating cellularity in subcortical
larity is assessed subjectively; otherwise, there is the possi-   regions, it is necessary to pay attention to bone structures
bility of computerized image analysis or histomorphometry.        in the biopsy (e.g., a tangentially taken sample with plenty
Bone marrow cellularity is demonstrated as the percentage         of cortical bone Fig. 1.3e). A representative biopsy (as
of a section occupied by hematopoietic tissue. This percent-      mentioned before) should show at least three intertrabecu-
age can either refer to the entire biopsy including bone tis-     lar spaces [9, 10].
1   Normal Bone Marrow                                                                                                                              7
b c
d e
Fig. 1.3 (a) Schematic representation of cellularity in normal bone          tical fatty tissue) (MGG). (c, d) Physiological subcortical hypoplasia: a
marrow biopsies showing decrease of hematopoiesis with advanced              common diagnostic pitfall (MGG). (e) The whole length of the biopsy
age. (b) BMB of a patient with myeloproliferative neoplasm (left side is     shows cortical bone (tangentially taken sample) (MGG)
representative for diagnosis, the biopsy on the right side reveals subcor-
8                                                                                                                     1   Normal Bone Marrow
1.2.1     Topography (Fig. 1.4)                                          one hand, “myeloid” implies the complete hematopoiesis, on
                                                                          the other hand this term describes the granulopoietic and
In the marrow cavities, hematopoietic tissue is distributed               monocytopoietic lineage only, as demonstrated by the
interstitial in the extravascular compartment (between the                myeloid: erythroid (M:E) ratio. Usually, there is no interpre-
sinusoids) (Fig. 1.4a, b). In some pathological conditions,               tation problem, but ambiguousness in using the term
hematopoiesis can be found intrasinusoidal also. Myeloid                  “myeloid” should be avoided.
precursor cells are closely attached to the endosteal surface                 Groups of erythropoietic cells and megakaryocytes show
and arterioles (Fig. 1.4c–f), more mature granulopoietic cells            a close association to the marrow sinusoids [3, 5, 8]
are seen in central intertrabecular areas. Attention should be            (Fig. 1.4g, h). In normal bone marrow, considerable varia-
paid to the term “myeloid,” which, in the German- and                     tions in the qualitative as well as quantitative distribution of
English-speaking parts, has two different meanings. On the                cellular components can be observed.
a b
c d
Fig. 1.4 (a, b) Distribution of hematopoiesis in the extravascular com-   around arterioles (MGG). (g) Erythropoietic island closely attached to
partment (MGG). (c, d) Myeloid precursors are located closely to the      sinus (MGG). (h) Close association of megakaryocyte to sinus (MGG)
surface of bone trabeculae (MGG). (e, f) Myeloid precursors located
1       Normal Bone Marrow       9
e f
g h
a b
Fig. 1.5 (a) Erythron with enclosed macrophage (mother cell) contain-        shows staining with antibody to Glycophorin C). (g, h) Normal BMB of
ing hemosiderin (Prussian blue). (b–d) Erythropoietic islands are nor-       older patient stained with H&E. On the right side, the same biopsy is
mally situated close to sinusoids (MGG). (e, f) Comparison of                immunohistochemically stained with the proliferation marker Ki67:
transferrin expression and glycophorin C expression in the same area of      Erythropoietic cells show the highest proliferation fraction
biopsy (left side: stained with antibody to transferrin [CD71], right side
1       Normal Bone Marrow           11
c d
e f
CD71 GlycC
g h
1.3.3     Granulocytopoiesis (Figs. 1.6 and 1.7)                       elocytes, and a rather weak positivity in more mature forms
                                                                        (Fig. 1.6e, f), whereas CD15 is strongly positive in metamy-
Sections of normal BMBs contain neutrophils, eosinophils,               elocytes and granulocytes (Fig. 1.7a–d) [19]. Neutrophils
and basophils, which can be identified by metachromatic                 and eosinophils are easily detectable in conventionally
stains (e.g., MGG) or immunohistochemically (Fig. 1.6a, b).             stained sections; however, for the detection of basophilic dif-
The granulocytopoiesis, representing 50–70% of nucleated                ferentiated cells, special stains, or immunohistochemical
cells, quantitatively dominates the hematopoiesis. According            investigations are necessary.
to maturation, myeloblasts, promyelocytes, myelocytes,                      If the myeloid:erythroid ratio is increased without
metamyelocytes, and segmented forms can be detected. The                decrease in erythroid cells, granulocytic hyperplasia is indi-
most immature myeloid cell detectable in bone marrow sec-               cated. In such conditions, either normal proportions of the
tions is the myeloblast, mostly located closely attached to the         various maturation stages are seen, or there can be a “shift to
trabecular surface, or periarteriolar (Fig. 1.6c, d). In greater        the left” with preponderance of immature forms, or a “shift
number promyelocytes and myelocytes are seen, which, in                 to the right” with an increase of mature granulocytes. As
thin stained sections show a granulation of the cytoplasm.              mentioned before, the paratrabecular and periarteriolar areas
These cells are located also in the generation zone of myeloid          constitute the generation zones of granulopoietic cells, more
cells. The more mature granulopoietic cells do not show any             mature forms are seen in central areas of marrow spaces.
connections with the paratrabecular or periarteriolar areas.            However, single immature granulopoietic cells are found
The distribution of granulopoietic cells can be highlighted             without recognizable connection to paratrabecular or periar-
immunohistochemically with antibodies to myeloperoxidase                teriolar areas. Normally, since granulocytopoiesis is very
(MPO) and CD15. MPO shows a strong positivity in promy-                 effective, all cells produced reach the circulation [4].
a b
c d
Fig. 1.6 (a) Eosinophils are easily identified with MGG. (b) A few      and periarteriolar (right side) (MGG). (e, f) Strong positivity of precur-
basophils are detected in normal bone marrow immunohistochemically      sor myeloid cells using an antibody to MPO
(2D7). (c, d) Myeloid precursors located peritrabecular (left biopsy)
1       Normal Bone Marrow                                                                                                                13
e f
MPO MPO
a b
CD15 MPO
c d
CD15 MPO
Fig. 1.7 (a–d) Normal bone marrow: Comparison of staining reactivity in myeloid cells using antibodies to CD15 (figures on the left side) and
MPO (figures on the right side)
14                                                                                                                 1   Normal Bone Marrow
1.3.4    Monocytopoiesis and Macrophages                             cells, or lipid. Occasionally, especially when there is an
          (Fig. 1.8)                                                  increased turnover of hematopoiesis, sea-blue histiocytes are
                                                                      found. These cells are macrophages with huge sea-blue cyto-
Even in optimal prepared histological sections, monocytes can         plasm corresponding to granules containing lipofuscin or
hardly be detected. These cells are easily mistaken for granulo-      ceroid (Fig. 1.8g, h). Quite often aggregates of sea-blue histio-
poietic precursor cells with their slightly kidney-shaped vesicu-   cytes are seen in myeloproliferative neoplasms and myelodys-
lar nuclei and eosinophilic variably granulated cytoplasm.            plastic syndromes [21, 22].
Small numbers of monocytes are located in the midst of hema-              Interstitial histiocytosis can occur due to an increased
topoietic islands or periarteriolar. Immunohistochemically,           turnover (sea-blue histiocytes) in the course of storage disor-
monocytes can be uncovered using antibodies to CD11c or               ders (congenital, kappa-positive crystal storage).
CD14 [20, 21] (Fig. 1.8a, b). In the bone marrow, monocytes               Increased histiocytes can be found in infectious disease
mature and become macrophages, which are found in great               (e.g., Mb. Whipple, atypical mycobacteriosis).
numbers diffusely dispersed (Fig. 1.8c–e). Macrophages often              A marked increase in histiocytes can be seen during resto-
contain hemosiderin (Fig. 1.8f), nuclear debris, hematopoietic        ration after myeloablative therapy.
a b
CD14 CD14
c d
CD163
Fig. 1.8 (a, b) Monocytes in normal bone marrow highlighted immu-     CD68. (f) Macrophages containing hemosiderin (Berlin blue iron
nohistochemically with an antibody to CD14. (c, d) Many macrophages   stain). (g, h) Aggregates of sea-blue histiocytes (in a patient with MDS)
(stained with an antibody to CD163) in normal bone marrow. (e)        stained with MGG
Macrophages in normal bone marrow stained with an antibody to
1       Normal Bone Marrow       15
e f
CD68
g h
1.3.5     Megakaryocytopoiesis (Figs. 1.9                               20–80 μm, their cytoplasm is less basophilic and often peri-
           and 1.10)                                                     nuclear granules can be detected. (c) Mature megakaryo-
                                                                         cytes with a diameter up to 150 μm exhibit an eosinophilic
Megakaryocytes with a diameter of 12 to 150 μm are the                   cytoplasm and show a variable granularity [4]. Especially
largest of bone marrow cells with an abundant cytoplasm                  when megakaryocytes are increased, emperipolesis
and lobulated nuclei (Fig. 1.9a–c). For detection of small               (pseudo-phagocytosis), the presence of other cells (lym-
forms, immunohistochemical stains are required. The pref-                phocytes, erythroblasts, erythrocytes, or granulocytes)
erential localization is near sinusoids (Fig. 1.9c, d).                  within megakaryocytes, is obvious [23] (Fig. 1.9g, h). This
However, since the sinusoids are often collapsed and thus                emperipolesis however, is without pathological signifi-
hardly apparent, megakaryocytes often seem to be diffusely               cance and can be seen in megakaryocytes of any size.
distributed within the marrow spaces. In normal hemato-                  Occasionally, denuded megakaryocyte nuclei are seen in
poiesis, usually megakaryocytes do not form clusters, but                normal bone marrows. Normal routinely stained bone mar-
are either seated singly or form loose cell groups counting              row sections reveal 8–15 megakaryocytes per mm2 corre-
up to 3–5 cells (Fig. 1.9e, f). During maturation, three                 sponding to 2–5 megakaryocytes per HPF [9]. Using
stages can be recognized. (a) Megakaryoblasts with a size                immunohistochemistry (e.g., CD41, CD42b, CD61) con-
of 12–20 μm show a kidney shaped or an oval nucleus with                 siderably more megakaryocytes, especially small ones, are
a basophilic cytoplasm. (b) Pro-megakaryocytes measure                   visible (up to 25/mm2) [5] (Fig. 1.10a–d).
a b
c d
Fig. 1.9 (a, b) Megakaryocytes with lobulated nuclei, the largest bone   ocytes in a patient with ITP. MGG. (g, h) Megakaryocytes with
marrow cells, are easily detected. MGG. (c, d) Normal perisinusoidal     emperipolesis. MGG
localization of megakaryocyte. MGG. (e, f) Loose groups of megakary-
1       Normal Bone Marrow       17
e f
g h
a b
CD41
c d
CD42b CD61
Fig. 1.10 (a) BMP of patient with untreated CML: In H&E stained biopsy, few small megakaryocytes are detectable. (b–d) Same biopsy as (a):
Immunohistochemically, using antibodies for megakaryocytes, much more megakaryocytes are visible: (b): CD41; (c): CD42b; (d): CD61
1   Normal Bone Marrow                                                                                                               19
1.3.6    Lymphocytes (Figs. 1.11 and 1.12)                            c arcinoma, cirrhosis, diabetes mellitus, systemic Castleman’s
                                                                        disease and drug therapy [24–26]. Nodular B- and T-cell aggre-
Lymphocytes are a component of the normal bone marrow pop-              gates increase with age and after medication (Fig. 1.12a, b).
ulation and appear either diffusely dispersed in the interstitium,          Whereas in the normal bone marrow of children the num-
or in the form of small aggregates or lymphoid nodules                  ber of lymphocytes is rather high with 30–60% (B-cells
(Fig. 1.11a–c). Lymphoid nodules or aggregates, especially if           comprise about 65% of lymphocytic cells in the first 4 years
they are rather small, are easily identified in sections stained for    of life), in adults the number of lymphocytes is much lower
reticulin fibers (e.g., Gomori’s stain), since these lymphoid           with 10–20%. Normally in adulthood, T-lymphocytes
aggregates show more fibers than the surrounding hematopoie-            (CD3+) outnumber B-lymphocytes (CD20+) (Fig. 1.12c, d).
sis. With increasing age lymphoid nodules occur more frequent           The rate of T-lymphocytes: B-lymphocytes is 4:1 to 6:1 [27].
and may cause differential diagnostic problems whether these            The ratio of CD 4 to CD8 cells of approximately 1:2 is the
lymphoid nodules are benign or malignant. In most cases, the            reverse of the ratio of CD4 to CD8 of 2:1 present in lymph
combination of adequate, representative, and good quality bone          nodes (Fig.1.12 CD4, CD8). Furthermore, CD8+ cells are
marrow t rephine biopsy, careful analysis of the cytomorphology        more frequent in the bone marrow than in peripheral blood.
of the lymphoid cells, number and pattern of their distribution,        An increase of CD8 is skewed in virus infection and autoim-
immunophenotype          based      on     immunohistochemistry         munity, CD4 is skewed in drug reactions. NK cells constitute
(Fig. 1.11d–h), and clinical and laboratory data allow the correct      about 2–4%.
interpretation to be made. In a few cases, cytogenetic and                  A reactive increase of T-cells with formation of lymphoid
molecular studies are needed for a correct diagnosis.                   aggregates is common (up to 40%) in elderly patients. An
    It should also be stressed that neoplastic BM involvement           increase of reactive T-cells is quite common in patients with
by hematolymphoid neoplasms may incite reactive inflam-                 MPN and MDS. In younger people, an increase of T-cells is
matory changes such as benign lymphoid aggregates, a dif-               found in viral infections and autoimmune disorders. Clonality
fuse increase in reactive mainly T-lymphocytes, plasma cells,           analyses can be misleading because of the presence of benign
histiocytes, and granuloma formation—the result of cytokine             reactive T-cell clones.
production by the lymphoma cells.                                           Furthermore, a reactive lymphocytosis can occur in the
    Another result of BM involvement by small cell lymphomas            form of hematogones after chemotherapy or post infectious.
is to cause reactive dysplastic features in the erythroid, granulo-         Characteristics of benign lymphoid aggregates of small
cytic and megakaryocytic lineages (reactive dyshematopoiesis).          size (<1 mm) with well defined borders are:
This secondary reactive myelodysplasia may mimic a primary              1. Random distribution; often perivascular and distant from bone
myelodysplastic syndrome (see Chapter - Myelodysplasia).                    trabeculae (lymphoid aggregates located paratrabecular are
    Lymphoid nodules are either well demarcated or with irreg-              always suspicious of infiltration of malignant lymphoma).
ular borders and may comprise germinal centers. Reactive lym-           2. Sometimes lymphoid follicles with germinal centers.
phoid infiltrates occur in a wide variety of diseases. These are        3. No cellular atypia.
infectious diseases such as hepatitis and HIV infection; con-           4. Predominance of B-lymphocytes, except in HIV
nective tissue disorders, particularly rheumatoid arthritis;                infection.
hematological neoplasia including lymphoma, MDS, and                    5. If plasma cells are found within lymphoid nodules, these
MPNs as well as idiopathic thrombocytopenic purpura, and a                  are polyclonal.
variety of anemias especially aplastic anemia, metastatic               6. Few reticulin fibers, except in HIV infection.
a b
Fig. 1.11 (a, b) Low and high magnification of lymphoid nodule: H&E. (c–h) Reactive lymphoid nodule with germinal center: (c): H&E; (d):
CD10; (e): CD20; (f): CD23; (g): CD3; (h): Ki67
20                             1   Normal Bone Marrow
c d
CD10
e f
CD20 CD23
g h
CD3 MIB1
a b
CD20
c d
    CD3                                                          CD3
    e                                                           f
CD4 CD8
Fig. 1.12 (a) Normal bone marrow: H&E. (b) Small number of B-lymphocytes in normal bone marrow (CD20). (c, d) Higher number of
T-lymphocytes in normal bone marrow (CD3). (e, f) Low number of CD4+ lymphocytes. Much higher number of CD8+ lymphocytes
22                                                                                                      1   Normal Bone Marrow
a b
c d
e f
try
Fig. 1.13 (a, b) Plasma cells with typical cartwheel cytoplasm around      red granules in the cytoplasm. MGG. (f) Mast cells in normal bone mar-
a vessel. MGG. (c, d) Mast cells located close to sinusoidal endothelial   row. (IH: tryptase)
cells. MGG. (e) High magnification of mast cells with densely packed
24                                                                                                                    1   Normal Bone Marrow
1.3.9    Bone Marrow Stroma (Fig. 1.14)                               2. Bone structure (architecture, osteopenia, osteosclerosis,
                                                                          increased remodeling)
Hematopoietic cells are embedded in a framework con-                   3. Cellularity (with consideration of the patients age: nor-
sisting of fat cells, fibroblasts, reticular cells, and the net-          mocellular, or mild, moderate, high hypocellularity or
work of blood vessels and sinusoids, which are often                      hypercellularity. Ideally, percentage of cellular compo-
collapsed and difficult to identify. Rarely nerves are                    nents and fatty tissue should be specified)
detected in marrow spaces (Fig. 1.14a, b). In special stains           4. Hematopoietic cells (erythro-, granulo-, megakaryocyto-
(e.g., Gomori’s stain), few fibers are seen in the normal                 poiesis: description of quantity, quality, and topography)
marrow, mostly around blood vessels and endosteum                      5. Stroma: fibroblasts, reticulin and collagen fibers, fat,
(Fig. 1.14c, d). This microenvironment supports the extra-                iron, blood vessels, stromal cells
vascular hematopoietic compartment [3, 8]. The content                 6. Other cellular components: B- and T-lymphocytes,
of iron, either within macrophages or in the bone marrow                  plasma cells, mast cells, macrophages, neoplastic infiltra-
stroma, can be graduated from 0 (no iron detectable) to 6                 tion, neoplastic hematological disease, (special reactive
(masses of clumped iron and extracellular iron) [33]                      changes).
(Fig. 1.14e–h).
   “Check list” for diagnosis of bone marrow biopsies:                 Diagnosis (ideally with consideration of clinical data): either
1. Assessment of length and quality (e.g., excellent, suffi-           descriptive (with statement of differential diagnoses) or
   cient, not adequate)                                                definitive (according to WHO criteria).
a b
c d
Fig. 1.14 (a, b) Nerve accompanying blood vessel (a: MGG, b:           detectable (grade 0). (f–h) Increase of iron (BBL): according to Gale:
Gomori’s stain). (c, d) Fibers around vessels in normal bone marrow    (f): grade 4, (g): grade 5. (h): grade 6 with very large deposits of intra-
(Gomori’s stain). (e) Using an iron stain (BBL) no iron deposits are   and extracellular iron
1       Normal Bone Marrow       25
e f
g h
2.2       Acquired Anemias                                              bone marrow biopsy, usually increased storage iron is
                                                                         detected. It is important to differentiate anemia due to
Anemia is the most common blood disorder with various                    iron deficiency from that of ACD since the treatment
causes. In the following chapter, various causes of anemia               options are different. Several tests differentiate iron defi-
are described.                                                           ciency anemia from ACD; however, the determination of
                                                                         iron stores by bone marrow examination remains the best
                                                                         method [3].
2.2.1     Anemia of Chronic Disease (ACD)
                                                                         2.2.1.1 Caution
Anemia of chronic disease (ACD) is the most common nor-                  In most cases, the underlying disease of anemia is unknown
mocytic anemia and the second most common form of ane-                   to the investigating pathologist [4]. Thus, in most cases only
mia worldwide (after iron deficiency anemia). ACD, also                  an exact descriptive diagnosis is possible.
called the anemia of inflammation, anemia of chronic                         Usually, patients with ACD are older than 60 years. A
inflammation, iron-reutilization anemia, or hypoferremia of              bone marrow biopsy differentiates between ACD and myelo-
inflammation was thought to be associated with infectious,               dysplasia, which is suspected in patients, who do not respond
inflammatory, or neoplastic diseases (Fig. 2.1a, b), but it has          to treatment.
been demonstrated that ACD can be seen in a variety of                       The bone in every bone marrow biopsy should be investi-
other conditions, such as severe trauma, autoimmune dis-                 gated carefully. The structure or changes of the bone in
eases like lupus or rheumatoid arthritis (Fig. 2.1c, d), diabe-          patients with anemia might be the key to the diagnosis. For
tes mellitus, HIV infection, kidney disease (Fig. 2.1e, f), and          example in patients with kidney problems, characteristic
anemia of older adults. This type of anemia is typically nor-            signs of renal osteopathy with a secondary hyperparathy-
mochromic, normocytic, hypoproliferative, and quite mild                 roidism showing the various amount of malacious compo-
in degree.                                                               nent or fibro-osteoclastic lesions can be detected. According
   The pathologic processes involved with ACD are                        to Delling [5], renal osteopathies can be categorized in three
decreased erythrocyte survival time, hindered erythropoi-                main types: (1) with marked fibro-osteoclastic lesions; (2)
esis, a reduced erythropoietin response, and pathologic                  with accentuated malicious component; and (3) combination
iron homeostasis [2]. A bone marrow biopsy is performed                  of (1) and (2) with (a) mild, (b) moderate, or (c) marked bone
if no underlying cause of the anemia is evident. In the                  remodeling with or without osteopenia.
a b
Fig. 2.1 Bone marrow of a 65-year-old female. The patient had been       to exclude myelodysplasia. The bone marrow shows moderate hyper-
treated for a diffuse large B cell lymphoma 8 months previously. The     cellularity (c) without dysplastic features. There is a slight increase in
patient has mild anemia, bone marrow biopsy was performed to exclude     iron (d). A 70-year-old woman was admitted because of moderate ane-
myelodysplasia. Bone marrow is slightly hypercellular for age (a), no    mia. The normocellular bone marrow (e) shows fibro-osteoclastic
dysplastic changes are evident. Increase of iron is seen (b BBL). Bone   lesions (f) and hyperparathyroidism was suggested by the pathologist.
marrow biopsy of a 69-year-old male with rheumatoid arthritis for        In this patient, an adenoma of the parathyroid gland was detected and
years. The patient has mild anemia and bone marrow biopsy was done       surgically removed
2       Non-neoplastic Conditions with Quantitative and Qualitative Changes in Hematopoiesis   29
c d
e f
2.2.2     Iron Deficiency Anemias (IDA)                                203 G/L; in differential count neutrophils were low with
                                                                        46%. Hepatitis C was verified (chronic hepatitis C, Genotype
Iron deficiency is prevalent. Although a bone marrow biopsy             3a). Mild hepatosplenomegaly was found. To exclude a
is not necessary in clinically straightforward IDA patients,            coexisting disease, a bone marrow biopsy was performed:
expected findings in most cases show hyperplasia of the ery-               The bone marrow was markedly hypercellular for age (no
throid series with rather small cells and the absence of stor-          fat cells were seen), due to a marked increase of erythroid
age iron and erythroid iron.                                            cells. The erythroid cells were arranged in large sheets with
   The differential diagnoses of IDA are numerous, including            mostly small forms. Iron could not be detected. In addition,
thalassemia, ACD and lead poisoning, and rare congenital                multifocal nests of histiocytic cells with strong PAS-
conditions, like congenital sideroblastic anemia and congeni-           positivity were seen. The diagnosis was descriptive with
tal atransferrinemia [6]. Thalassemia disorders, hereditary             hypercellular bone marrow, increased erythroid cells, lack of
diseases, are various and result from reduced globin chain              iron staining and groups of PAS-positive histiocytes
synthesis. The disorder results in excessive destruction of red         (Fig. 2.2a–h). Although the patient had no clinical signs for a
blood cells, which leads to anemia. The resulting microcytic            storage disease or Whipple’s disease, the possibility of such
and hypochromic red blood cell counts arise from inadequate             a disease was mentioned in a commentary.
availability of either alpha- or beta-globin chains for hemo-
globin production. Since thalassemia is one of the most com-  2.2.2.1 Caution
mon genetic disorders worldwide, the bone marrow biopsy of    The evaluation of iron in decalcified bone marrow biopsies
a patient with thalassemia minor is shown (Fig. 2.2a–h).      might be biased by the decalcification process when iron can
                                                              be eluted. If possible, bone marrow aspirates should be
Case History of Patient with Thalassemia Minor                examined for iron, which is considered the gold standard in
A 24-year-old male was admitted to the hospital with a sus- evaluating iron-depleted states. The absence of bone marrow
pected diagnosis of hepatitis C. The patient had anemia and iron stores is not necessarily predictive of IDA. Such a find-
known thalassemia intermedia (homozygotic beta-ing needs to be evaluated in conjunction with other labora-
thalassemia, beta-plus variant). Since the age of 3 years, he tory findings and the clinical findings since certain other
received several blood transfusions, the last one 6 months hematological diseases may coexist [7].
previously. His blood cell count at admission was: Leuko         If macrophages/histiocytes are markedly increased, this
10.2 G/L, Ery 4.61 T/L, Hb 8.1 G/DL (low), Hkt 26.8/L % should be mentioned in the report and differential diagnoses
(low), MCV 58.1/L FL (low), MCH 17.6 PG (low), MCHC have to be listed (in this special case, the clinicians should
30.2 G/DL (low), RDW-CV 33.4% (high), Thrombo exclude storage disease or Whipple’s disease).
a b
Fig. 2.2 Thalassemia. Highly hypercellular bone marrow of a             vacuolated cytoplasm. (f) Sheets of PAS-positive macrophages. (g)
24-year-old male (a, H&E) due to the increase in erythroid cells (b,    Granulocytopoiesis is slightly decreased (immunohistochemical stain
H&E). (c) Erythroid cells are immunohistochemically stained             with an antibody to CD15). (h) Normal number of rather small
(Glycophorin A). (d) Iron stain did not show iron deposits (BBL). (e)   CD42b-positive megakaryocytes
Groups of macrophages/histiocytes with pale eosinophilic, slightly
2       Non-neoplastic Conditions with Quantitative and Qualitative Changes in Hematopoiesis       31
c d
GlycA BBL
e f
PAS
g h
CD15 CD42b
2.2.3     Megaloblastic Anemias                                            nucleus with often rod-shaped nucleolus. Concerning the
                                                                            myeloid series, the most striking features are the occurrence of
Megaloblastic anemia is a condition in which unusually                      nuclei with hypersegmentation and/or gigantism of bands.
large, structurally abnormal, immature red blood cells (meg-
aloblasts) can be found in the bone marrow. The most com-                   Case History
mon causes of megaloblastic anemia are deficiency of either                 A 23-year-old female was admitted because of fatigue, fever
vitamin B12 or folate (vitamin B9). Furthermore, megalo-                    of unknown origin and marked megaloblastic anemia. Blood
blastic anemia of diverse etiology is a common complication                 picture showed Leuko 8.85 G/L, Ery 1.86 T/L, Hb 6.9 g/dL,
of chronic liver diseases: the causes include acute or chronic              Hkt 19.5%, MCV 104.8 fL, MCH 37.1 pg, MCHC 35.4 g/
gastro-intestinal hemorrhage, and hypersplenism secondary                   dL, Thrombo 210/L. A bone marrow biopsy was performed
to portal hypertension. Splenomegaly, which is usually                      and megaloblastic anemia was diagnosed (Fig. 2.3a–h). In
caused by portal hypertension in patients with chronic liver                the meantime, the level of vitamin B12 was done which was
disease, may lead to secondary hemolysis, an increase in                    markedly decreased.
plasma volume and megaloblastic anemia. Alcohol is a fre-
quent etiologic factor of chronic liver disease. Alcoholics                 2.2.3.1 Caution
often develop secondary malnutrition, a manifestation of                    Megaloblastic anemia with typical erythroid lineage abnor-
which may be anemia caused by folic acid deficiency [8].                    malities may be masked by concurrent iron deficiency.
Without these vitamins, the synthesis of deoxyribonucleic                      Occasionally, the bone marrow is densely packed with
acid (DNA) is hampered. Megaloblastic anemias are either                    megaloblasts. Such marrows morphologically might be mis-
constitutional or acquired, which are much more common.                     diagnosed (prior to immunohistochemical investigations) as
Even megaloblastic anemias in children are more often                       acute leukemias. To avoid this misdiagnosis, erythropoietic
caused by dietary factors than constitutional. The spectrum                 markers should be used. In this context, it is important to
of etiologies associated with macrocytic anemia includes                    state that CD71 (transferrin receptor) is expressed at high
furthermore drugs, neoplastic hematologic diseases like                     levels in erythroblasts at all stages of development, but is
myelodysplasia, plasma cell myeloma and leukemia, alco-                     absent from the majority of mature cells. This staining pat-
holism, and other chronic illnesses (e.g., hypothyroidism).                 tern greatly facilitates the detection of cells of erythroid lin-
   The bone marrow of patients with megaloblastic anemia is                 eage in bone marrow biopsy specimens. However, be aware,
typically hypercellular with marked hyperplasia of the ery-                 that CD71 is very weakly expressed (or even missing) in
throid cells (Fig. 2.3, case history) but in most cases also of             erythroleukemia (which is very rare); furthermore, CD71
granulopoietic cells. Usually, many mitoses can be found. The               can be expressed in cases of acute myeloid leukemias and
typical megaloblast in the bone marrow shows an enlarged                    acute lymphatic (B and T) leukemias.
a b
Fig. 2.3 Megaloblastic anemia. (a) Overview of the bone marrow              locytes are present. (e) No fibrosis is seen in these highly hypercellular
biopsy (H&E) with highly hypercellular marrow spaces for the age of         bone marrow (Gomori’s stain). (f) Erythropoiesis, stained with an anti-
23. (b) In the center, a typical megaloblast with a rod-like nucleolus is   body to glycophorin C, is increased. (g) Immature granulopoietic cells
visible (H&E). (c) Megaloblasts are nicely recognized in MGG stained        are regularly located paratrabecular or around vessels. (MPO). (h)
sections. (d) In the center of the marrow spaces, coarse rod-like granu-    Normal appearing megakaryocytes (CD42b)
2       Non-neoplastic Conditions with Quantitative and Qualitative Changes in Hematopoiesis       33
c d
MGG MGG
e f
GOM GlycC
g h
MPO CD42b
2.2.4     Hemolytic Anemia                                               gene mutation [11]. The paroxysmal nocturnal hemoglobin-
                                                                          uria-phenotype granulocytes can be assessed by using mul-
In hemolytic anemias, the red blood cells are destroyed before            tiparameter flow cytometry immunophenotypic analysis
their life span is over. In cases of extrinsic hemolytic anemia,          with antibodies specific for glycosylphosphatidylinositol-
known as autoimmune hemolytic anemia, a bone marrow                       anchored proteins (CD55, CD59, CD16, CD66b). For the
biopsy is performed to exclude an underlying hematologic                  investigating hematopathologist, the knowledge of existing
neoplasm. There are several diseases and even medications                 PNH clones is essential for making a correct diagnosis.
that can cause such a condition. Some underlying causes of
extrinsic hemolytic anemia are splenomegaly, drugs, lupus,                Case History
hepatitis, infection with EBV, but also leukemias, lymphomas,             A 63-year-old female with hemolytic anemia and verified
myeloproliferative neoplasms [9], and solid tumors.                       PNH underwent a bone marrow biopsy (Fig. 2.4a–h) for
   Among the acquired hemolytic anemias, paroxysmal                       exclusion of a concomitant disease. The information, given
nocturnal hemoglobinuria (PNH) is of special interest. PNH                by the clinicians was reticulocytosis, high concentration of
can be associated with aplasia and pancytopenia, imitating                LDH and indirect bilirubin, and abnormally low concentra-
aplastic anemia, but usually the bone marrow shows a                      tion of serum haptoglobin. The bone marrow was hypercel-
marked increase of nucleated erythroid precursors without                 lular mainly caused by an increased erythropoiesis. No
dysplastic features and a reversed myeloid to erythroid ratio             dysplastic changes could be detected. A descriptive diagno-
(Fig. 2.4, case history) [10]. PNH is a clonal hematopoietic              sis consistent with hemolysis and PNH was performed.
stem cell disease, caused by mutations in the PIG-A gene
[11]. An increased number of cells with deficiency of glyco-              2.2.4.1 Caution
sylphosphatidylinositol (GPI)-anchored membrane proteins                  Based upon morphology and also immunohistochemistry, a
are a result of hematopoietic progenitor cells with a phos-               diagnosis of PNH cannot be made. Clinical information
phatidylinositol glycan complementation group A (PIG-A)                   about the presence of a PNH clone is mandatory.
a b
Fig. 2.4 PNH. (a, b) Routinely stained H&E section shows marked           enlarged erythropoietic cell nests with a slight left shift. No dysplastic
hypercellular bone marrow in this 63-year-old female due to an increase   changes are evident. (g) Megakaryocytes, stained with an antibody to
of erythropoietic cells. Erythropoiesis is stained with antibodies to     CD42b are slightly decreased and small. (h) Myelopoiesis, stained with
CD71 (c, d), Glycophorin C (e) and Glycophorin A (f). There are rather    an antibody to MPO appears normal
2       Non-neoplastic Conditions with Quantitative and Qualitative Changes in Hematopoiesis       35
c d
CD71 CD71
e f
GlycC GlycA
g h
CD42b MPO
a b
Fig. 2.5 Post-hepatitic SAA. (a) The bone marrow is highly hypocel- an antibody to CD71. (d) Only a few diffusely distributed myeloid cells
lular for age (MGG). (b) Occasional erythropoietic islands, the so- can be seen (MPO). (e) No fibrosis is seen (Gomori’s stain). (f) Slight
called hot spots, are seen in higher magnification (MGG). No increase of iron deposits in the bone marrow (BBL)
megakaryocytes are visible. (c) Erythropoietic cells are visualized with
2       Non-neoplastic Conditions with Quantitative and Qualitative Changes in Hematopoiesis     37
c d
CD71 MPO
e f
GOM BBL
2.2.5.1 Caution                                                          aplasia (Fig. 2.6a–f), pure white cell aplasia (T-LGLL, idio-
A distinction between aplastic anemia and hypoplastic                     pathic), and thrombocytopenia due to amegakaryocytosis.
myelodysplasia, especially in the elderly, can be                         Pure red cell aplasia in adults is diagnosed when isolated
problematic. Dyserythropoiesis can occur in aplastic ane-                anemia, in the presence of normal white cell and platelet
 mia, however, megakaryocytes, if present, are not atypical               counts, is associated with a normocellular marrow with
 in aplastic anemia. Furthermore, in aplastic anemia no                   nearly complete absence of erythroblasts but normal myeloid
 abnormal neutrophils, no blasts (CD34) and no fibrosis are               cells and megakaryocytes (Fig. 2.6a–f).
 usually found.                                                              Pure red cell aplasia is occasionally associated with lym-
    In addition to hypoplastic myelodysplasia, differential               phomas (AITL, cHL, DLBCL, B-CLL, T-LGLL).
 diagnosis to aplastic anemia includes hypocellular acute                    Hepatitis-associated aplastic anemia is well known and
 lymphatic leukemia (Cave: the bone marrow of patients with               usually occurs 2–3 months after an acute episode of
 aplastic anemia often show an increase in lymphocytes and                hepatitis.
 mast cells), infections (especially mycobacterial infections),              Secondary aplasia of the erythroid cells (erythroblastope-
 malnutrition (gelatinous transformation of the bone marrow               nia) can also be caused during pregnancy, in patients with
 stroma), aplastic single-lineage cytopenias, like pure red cell          thymoma or due to anti-recombinant EPO-antibodies.
a b
c d
CD71 GlycA
Fig. 2.6 Pure red cell aplasia. (a, b) Normocellular bone marrow          A, e: glycophorin C) a highly decreased, nearly missing erythropoiesis
biopsy of a 50-year-old female with pure red cell aplasia due to a thy-   is evident. (f) Granulopoiesis is normal concerning quantity as well as
moma (H&E). Using erythropoietic markers (c: CD71, d: glycophorin         quality (myeloperoxidase)
2       Non-neoplastic Conditions with Quantitative and Qualitative Changes in Hematopoiesis     39
e f
GlycC MPO
a b
Fig. 2.7 CVD. (a, b) In routinely stained H&E section, the bone mar-    antibody to CD15. Only a few single CD15-positive cells are present.
row is moderately hypercellular. With antibodies to MPO (c) and CD33    (f) Normal appearing erythropoietic cells (antibody to CD71). A few
(d) myeloid precursor cells are seen. Occasionally, ALIPs can be        CD20-positive B cells are seen (g), there are many CD3-positive T-cells
detected. (e) Absence of mature granulocytes is demonstrated using an   diffusely dispersed (h)
2       Non-neoplastic Conditions with Quantitative and Qualitative Changes in Hematopoiesis      41
c d
MPO CD33
e f
CD15 CD71
g h
CD20 CD3
a b
c d
Fig. 2.8 SLE. (a, b) Routinely stained H&E section show moderately        myeloid cells as shown with an antibody to MPO. (f) Normoquantitative
hypocellular bone marrow with a nodular lymphocytic infiltrate. (c)       erythropoiesis with dysplastic and megaloblastic changes (erythropoi-
The lymphocytic infiltrate is composed of many CD3-positive T-cells       etic cells are stained with an antibody to glycophorin C). (g) There is no
(left) and many, centrally located CD20-positive B cells (right). (d)     increase of blast cells (immunohistochemical stain with an antibody to
Using antibodies to light chains kappa and lambda the B cells are poly-   CD34). (h) Megakaryocytes are normocellular but slightly atypical
clonal (kappa left side, lambda right side). (e) Marked decrease of       with rather small forms (antibody to CD42b)
2       Non-neoplastic Conditions with Quantitative and Qualitative Changes in Hematopoiesis       43
e f
MPO GlycC
g h
CD34 CD42b
a b
c d
e f
CD20 CD23
Fig. 2.9 Castleman’s disease. (a, b) In H&E stained section, the over-       cal for a reactive lymphoid follicle. (o) Most of the blasts are CD20-
view shows highly hypercellular bone marrow with multiple nodular            positive. (p) Many blasts show reactivity for CD30 (EBV done with
lymphoid infiltrates, most of which are closely attached to the bony         EBER, [not shown], is negative). (q, r) Using a plasma cell marker
trabeculae (b). (c, d) Some of the lymphoid infiltrates contain diffusely    (Vs38c) in the periphery of the lymphoid aggregates many plasma cells
dispersed blasts. Most lymphoid aggregates resemble reactive follicles       are evident. (s) With antibodies to light chains kappa (left) and lambda
with many CD20-positive B-cells (e), a network of follicular dendritic       (right) the plasma cells are polyclonal. Within the lymphoid aggregates
cells (f, CD23), many CD3-positive T-cells (g) and BCL2-negative fol-        the B lymphocytes do not show clonality. (t) There is no increase of
licle center (h). Lymphoid nodules stained with various T-cells markers      IgG4-positive plasma cells. (u) Granulopoiesis (MPO) is increased. (v)
show a distribution like in reactive follicles (i: CD4; j: CD5; k: CD7; l:   Erythropoietic cells (CD71) are increased. (w, x) There is an increase of
CD8). (m) Lymphoid nodule with many diffusely dispersed blasts               rather small not atypical megakaryocytes (CD42b)
(H&E). (n) The proliferation fraction (Ki67) is quite low and not typi-
46                     2   Non-neoplastic Conditions with Quantitative and Qualitative Changes in Hematopoiesis
g h
CD3 BCL2
i j
CD4 CD5
k l
CD7 CD8
m n
Ki67
o p
CD20 CD30
q r
Vs38c Vs38c
s t
u v
MPO CD71
w x
CD42b CD42b
2.5       Non-neoplastic Conditions                                with the presence of immature precursors and absence of
           with Quantitative and Qualitative                        mature granulopoiesis, the bone marrow may resemble pro-
           Changes in Granulopoiesis                                myelocytic leukemias.
a b
c d
MPO MPO
e f
CD42b CD71
Fig. 2.10 Agranulocytosis. The bone marrow is normo- to slightly   myeloid precursors (MPO). (e) Megakaryocytopoiesis is normal
hypercellular for age (a, H&E). In some areas, marked edematous    appearing (CD42b). (f) Erythropoietic cells are slightly increased
stroma with inflammatory cells is evident (b, H&E). (c, d)         (CD71)
Granulocytopoiesis is highly decreased with only some dispersed
2       Non-neoplastic Conditions with Quantitative and Qualitative Changes in Hematopoiesis                                               51
a b
c d
MPO CD71
Fig. 2.11 Kostmann disease. (a–d) These figures show the first bone      (d). A bone marrow biopsy was taken 6 months after GCSF therapy: In
marrow biopsy of the girl. In routinely stained H&E sections, the bone   routinely stained H&E sections (e, f), normocellular bone marrow is
marrow is normocellular for age but no mature granulopoiesis is seen     seen. Using an antibody to MPO immature granulopoietic cells are con-
(a, b). Using an antibody to MPO (c) the granulopoiesis is immature      centrated around vessels and trabeculae (g). Erythropoietic cells
with a marked maturation arrest. The erythropoiesis (CD71) is normal     (CD71) are not affected (h)
52                      2   Non-neoplastic Conditions with Quantitative and Qualitative Changes in Hematopoiesis
e f
g h
MPO CD71
2.7           Leukemoid Reaction (LR)                                  karyocytopoiesis appeared rather normal. A descriptive diag-
                                                                        nosis was made. Two weeks later, the WBC count decreased
In LR, a marked elevation in the WBC count resembling leuke-            to normal levels without intensive therapy. The reason for her
mia is found. The WBC count can exceed 50.00 × 109/L and is             increased WBC count could not be evaluated.
composed mainly of segmented neutrophils which may or may
not show toxic changes. The leukocytosis is associated with a
cause outside the bone marrow: this can be some sort of infec-          2.7.1     Caution
tious diseases, paraneoplastic reactions, injuries or hemor-
rhages and hemolytic events [27]. The bone marrow in LR is              LR is an important, probably the most important, differential
mildly to marked hypercellular caused by an increase of neu-            diagnosis in hematological malignancies since misinterpretation
trophilic myeloid hyperplasia and occasionally additional               of secondary leukemia or relapse of a leukemia is possible.
megakaryocytic hyperplasia (see Fig. 2.12, case history below).             Chronic neutrophilic leukemia, an extremely rare neo-
                                                                        plasm, has to be taken into consideration, if a patient has
Case History                                                            prolonged neutrophilia. Clonality of neutrophils thus has to
An 81-year-old woman, who has been treated for diffuse                  be demonstrated or excluded for differential diagnosis.
large B-cell lymphoma 6 years previously, was admitted with                 BCR-ABL-positive CML morphologically can resemble
a WBC of 62.50 × 109/L. Clinical workup including cytoge-               LR. However, in LR the megakaryocytes can be helpful for
netics did not reveal any evidence of malignant disease. A              differential diagnosis since in LR the megakaryocytes are
bone marrow biopsy was performed (Fig. 2.12a–d). This                   larger (normal looking) than in CML. Furthermore, in LR
biopsy was markedly hypercellular for age due to an increased           usually no increase of basophils is found.
granulopoiesis with maturation. Erythropoiesis and mega-                    LR can also occur as a rebound effect after neutropenia [28].
a b
MGG
c d
MPO
Fig. 2.12 Leukemoid reaction. In routinely stained H&E (a) and MGG (b) sections, the bone marrow is highly hypercellular for age. No neoplas-
tic infiltrate is seen. (c) The maturing granulopoiesis is highly increased (MPO). (d) Note the rather polymorphic megakaryocytopoiesis (MGG)
54                                          2   Non-neoplastic Conditions with Quantitative and Qualitative Changes in Hematopoiesis
2.8      Granulopoiesis with Increased                            dysplastic changes in multiple cell lines, it is more likely to
          Eosinophils                                              deal with a hematological malignancy.
Increased eosinophils in the blood (>0.5 × 109/L) are a fre-       Case History (Reactive Eosinophilia)
quent finding, and is in most cases connected with eosino-         A 68-year-old male was admitted with pruritus and dyspnea.
philia (>10%) in the bone marrow. Patients with mild               The patient is under observation for treated diffuse large
eosinophilia usually are asymptomatic, in cases with moder-        B-cell lymphoma 5 years previously. At admission, the patient
ate to marked eosinophilia there is a risk of severe organ         showed leucocytosis with eosinophilia and anemia. His blood
damage, irrespective of the cause of eosinophilia. To avert        count was:: Leuko 39.78 G/L, Ery 3.58 T/L, Hb 11.0 g/dL,
organ damage and fibrosis, which is caused by the release of       Hkt 31.2%, normal values of MCV, MCH, and MCHC,
special proteins from the eosinophilic granules, it is essential   Thrombo 271 G/L, MPV 9.5 fL; segmented 12% (normal val-
to identify the cause of eosinophilia as soon as possible.         ues 50–75), eosinophils 82% (normal: 5%), monocytes 3%
    An absolute eosinophilic count of more than 500/μL is          (normal), and lymphocytes 3% (normal 20–40). According to
defined as eosinophilia. An eosinophilia may occur either          the patient, he suffered from an influenza infection 2 weeks
as (1) reactive (secondary) eosinophilia, (2) clonal (neo-         ago. Cytogenetically and by molecular techniques, no abnor-
plastic) eosinophilia, and as (3) idiopathic eosinophilia, if      malities could be detected. The clinical workup included a
no underlying reactive cause is found and there is no evi-         bone marrow biopsy (Fig. 2.13a–h).
dence that the eosinophils are derived from a clonal myeloid          The tumor-free bone marrow biopsy was moderately
neoplasm.                                                          hypercellular for age. Granulocytopoiesis, which showed
    Various causal connections for reactive eosinophilia are       maturation, was increased and marked eosinophilia with
known [29]. Frequent causes for eosinophilia are infections        more than 80% eosinophils was detected in routinely stained
(especially helminths, Borrelia Burgdorferi, fungi), allergic      sections and immunohistochemically. The diagnosis was
reactions and asthma, respiratory diseases like Loeffler syn-      descriptive with a comment that a differential diagnosis
drome and hypersensitivity pneumonitis, cytokine infusions,        between primary and secondary eosinophilia is not possible
vasculitis, non-hematological malignant diseases, drug reac-       based on morphology.
tions, and connective tissue diseases. Furthermore, eosino-
philia can occur in combination with neoplastic disease,
especially Hodgkin’s disease, Langerhans cell histiocytosis,       2.8.1    Caution
and T-cell lymphomas, but also in combination with solid
tumors, especially in stomach and lung. If an unexplained          In many cases with eosinophilia, it may be impossible to dif-
eosinophilia of more than 1.500/μL persists for a longer           ferentiate primary from secondary eosinophilia. Thus, cyto-
period than 6 months, this eosinophilia is termed “hypereo-        genetic and molecular studies are essential.
sinophilia” and may be more likely primary than secondary.             In bone marrow biopsies with marked eosinophilia, an
The morphology of eosinophils does not differentiate               infiltration by malignant lymphoma (T-cell lymphoma,
between primary and secondary eosinophilia (Fig. 2.13, case        Hodgkin’s disease) has to be excluded. Since lymphoma
history). For an exact diagnosis, complex cytogenetic and          infiltration can be subtle, at least some immunohistochemi-
molecular abnormalities have to be investigated. If the bone       cal stains with B- and T-cell markers and CD30 should be
marrow with an increase of eosinophils shows atypia and            performed.
2       Non-neoplastic Conditions with Quantitative and Qualitative Changes in Hematopoiesis                                                55
a b
MGG
c d
EPX MPO
Fig. 2.13 Reactive eosinophilia. (a) The bone marrow is moderately       cells are stained. To exclude an infiltration by mast cells, which can
hypercellular. Even at low power magnification, an increase of eosino-   resemble eosinophils, mast cell markers CD117c, (e) and tryptase (f)
phils is seen (H&E). (b) Increased granulopoiesis with marked eosino-    were used. Only a few mast cells are evident. (g) Erythropoiesis is
philia (>80%) is verified using a metachromatic stain (MGG). (c) The     slightly decreased and left shifted, but not dysplastic (CD71). (h)
eosinophils are immunohistochemically stained with an antibody to        Megakaryocytes are rather small and slightly decreased, but character-
EPX. (d) Using an antibody to MPO, the more immature granulopoietic      istic atypias within megakaryocytes could not be detected (CD42b)
56                      2   Non-neoplastic Conditions with Quantitative and Qualitative Changes in Hematopoiesis
e f
CD117c Trypt
g h
CD71 CD42b
a b
MGG MGG
c d
MGG MGG
Fig. 2.14 Megakaryocytes (MGG) with emperipolesis in PMF (a) and      karyocyte within another megakaryocyte could be detected (MGG). (d)
ET (b). (c) In the bone marrow of a patient with MDS/MPN with ring    A bone marrow biopsy with dense infiltration by AML-blasts shows an
sideroblasts and thrombocytosis an unusual emperipolesis of a mega-   emperipolesis by a neoplastic blast (MGG)
58                                              2   Non-neoplastic Conditions with Quantitative and Qualitative Changes in Hematopoiesis
a b
MGG
c d
CD42b
Fig. 2.15 Increase of small megakaryocytes with often condensed          cytes (H&E). (f): immunohistochemical stain with CD61. (g, h)
chromatin are a characteristic finding in BCR-ABL-positive CML (a:       Increase of megakaryocytes with a left shift in a patient with ITP. (g:
H&E). Staining with MGG (b) reveals also an increase in basophils. (c)   H&E; h: MGG). (i, j) This bone marrow was taken after chemotherapy
In a patient with MDS with multilineage dysplasia small atypical mega-   for acute myeloid leukemia. In this bone marrow, a marked increase of
karyocytes with hypolobulated nuclei are seen (H&E). (d)                 rather small left shifted megakaryocytes is evident (i: H&E; j: immuno-
Immunohistochemically, much more small atypical megakaryocytes           histochemical stain with CD42b). (k, l) This bone marrow biopsy of an
can be found in the bone marrow of the same patient (see c) with MDS     old patient (staging was done for prostatic carcinoma) shows mega-
with multilineage dysplasia (CD42b). (e) Bone marrow biopsy with         karyocytes of normal size, but with hypolobulated nuclei (H&E)
MDS with isolated del(5q) reveals masses of small atypical megakaryo-
2       Non-neoplastic Conditions with Quantitative and Qualitative Changes in Hematopoiesis             59
e f
CD61
g h
MGG
i j
CD42b
k l
a b
MGG MGG
Fig. 2.16 (a, b) Bone marrow biopsy of a patient with ET showing huge megakaryocytes with hyperlobulated nuclei (MGG)
62                                             2   Non-neoplastic Conditions with Quantitative and Qualitative Changes in Hematopoiesis
a b
c d
CD42b
Fig. 2.17 Bone marrow biopsy in a patient with PMF. Even at low         In routinely stained H&E section within an edematous stroma, mega-
magnification in routinely stained H&E sections (a), the naked nuclei   karyocytes with naked nuclei are seen (c). Staining with an antibody to
of megakaryocytes are visible due to their dense dark chromatin. In     CD42b (d) highlights the megakaryocytes, some of which are denuded
higher magnification, the naked forms do not show a visible cytoplasm
(b). This bone marrow biopsy was taken from an HIV-positive patient.
2       Non-neoplastic Conditions with Quantitative and Qualitative Changes in Hematopoiesis                                                  63
a b
CD42b
c d
CD42b
Fig. 2.18 (a) Bone marrow biopsy of a 55-year-old male after chemo-      increase of small atypical megakaryocytes forming clusters and sheets
therapy of AML. The bone marrow is hypercellular with increased left     (CD42b). (i) Highly hypercellular bone marrow of a 63-year-old female
shifted hematopoiesis (H&E). (b) Clusters of rather small megakaryo-     diagnosed with MDS/MPN with ring sideroblasts and thrombocytosis
cytes after chemotherapy are visualized immunohistochemically            (MDS/MPN-RS-T) (H&E). (j) Increased highly atypical megakaryo-
(CD42b). (c) Highly hypercellular bone marrow of a 46-year-old male      cytes in this patient are forming clusters (CD42b). (k) Highly hypercel-
diagnosed with MDS with blast excess-2 (H&E). (d) Clusters of small      lular bone marrow of a 74-year-old female with MPN, early phase of
atypical megakaryocytes with hypolobulated nuclei are seen by immu-      PMF (pre-PMF) (H&E). (l) Clusters of large atypical megakaryocytes
nohistochemistry in this 46-year-old male with MDS with blast excess-2   in the patient with pre-PMF (CD61). (m) Highly hypercellular bone
(CD42b). (e) Normocellular marrow in a young 18-year-old male diag-      marrow of a 50-year-old male with MPN pre-PMF (H&E). (n) The
nosed with MDS with multilineage dysplasia (H&E). (f) Clusters and       increased atypical rather large megakaryocytes in the 50-year-old
even sheets of atypical small megakaryocytes are highlightened immu-     patient with pre-PMF are occasionally arranged in clusters (CD61). (o)
nohistochemically (CD42b). (g) Hypercellular bone marrow biopsy of       Hypercellular bone marrow in a 70-year-old male with MPN PMF
a 87-year-old male diagnosed with MDS with isolated del(5q) (H&E).       (fibrotic phase) (H&E). (p) Clusters of rather large atypical megakaryo-
(h) The hallmark of the bone marrow in this patient is the markedly      cytes are found in this 70-year-old male with PMF (CD61)
64                      2   Non-neoplastic Conditions with Quantitative and Qualitative Changes in Hematopoiesis
e f
CD42b
g h
CD42b
i j
CD42b
k l
CD61
m n
CD61
o p
CD61
2.14     Intrasinusoidal Megakaryocytes                              Especially, if the sinuses are dilated, megakaryocytes within
          (Fig. 2.19)                                                 these vessels easily can be recognized. Intrasinusoidal local-
                                                                      ization of megakaryocytes is not a feature of “normal” bone
Intrasinusoidal megakaryocytes usually are a feature of               marrows.
MPNs and of the rare autoimmune caused myelofibrosis [35].
a b
MGG MGG
c d
Fig. 2.19 (a, b) Intrasinusoidal megakaryocyte with hyperlobulated    (g) Intrasinusoidal megakaryocyte in a patient with PMF (MGG). (h)
nuclei typically for MPN, ET (MGG). (c, d) Low and high power mag-    Intrasinusoidal megakaryocytes in a patient with MPN. This bone mar-
nification of a case with MPN, PV. Even at low power magnification,   row morphologically resembles in one way MPN PV and in another
intrasinusoidal megakaryocytes are seen (H&E). (e) Huge megakaryo-    way PMF. Clinically, the patient has no signs of polyglobulia, periph-
cytes located within a sinus in MPN, ET (MGG). (f) Intrasinusoidal    eral blood shows slight thrombocythemia and leukocytosis. (MGG)
megakaryocytes in a patient with MPN, post-PV MF (H&E).
2       Non-neoplastic Conditions with Quantitative and Qualitative Changes in Hematopoiesis     67
e f
MGG
g h
MGG MGG
a b
                                                                  MGG                                                                   GOM
c
                                                                             d
GOM
Fig. 2.20 (a, b) Paratrabecular megakaryocytes in a 87-year-old male         lular bone marrow of a 67-year-old male with MDS/MPN with ring
patient with MDS with isolated del(5q). (left: MGG; right Gomori’s           sideroblasts and thrombocytosis (MDS/MPN-RS-T). Many megakary-
stain). (c, d) A 47-year-old patient with treatment-associated MDS with      ocytes are found closely attached to the bony trabeculae (e: H&E; f:
fibrosis (left: H&E, right: Gomori’s stain). (e, f, g, h) Highly hypercel-   CD42b; g, h: CD61)
2       Non-neoplastic Conditions with Quantitative and Qualitative Changes in Hematopoiesis       69
e f
CD42b
g h
CD61 CD61
2.16       Gray Platelet Syndrome                                             brother also had low platelet counts and died aged 33 due to
                                                                               spontaneous cerebral hemorrhage. Her parents, husband, and
Gray platelet syndrome (GPS) is a rare inherited thrombocy-                    the children of the deceased brother were clinically
topenia characterized by macrothrombocytopenia and the                         unaffected. Further studies showed that the respective family
absence of platelet α-granules resulting in typical gray plate-                 members are affected by gray platelet syndrome and a novel
lets on peripheral smears [37]. The disease is present at birth,                homozygous single nucleotide insertion in GFI1B
but occasionally the disease is discovered in adulthood.                        (NM_004188.5; c.551insG) could be detected in the affected
Among the 60 well-documented cases, no female-male gen-                        mother.
der inequality and no ethnic or geographic predisposition is                       The bone marrow biopsy of the 36-year-old woman
found. Patients with gray platelet syndrome are often diag-                     was slightly hypercellular with a slight increase of reticu-
nosed as immune thrombocytopenic purpura (ITP). Long-                           lin fibers. The most conspicuous finding was that of a
term follow-up data show the progressive nature of                              moderately increased megakaryocytopoiesis. The mega-
thrombocytopenia and myelofibrosis of GPS resulting in                          karyocytes were rather small with either normally lobu-
fatal hemorrhages in many patients.                                             lated or hypolobulated nuclei with condensed chromatin.
                                                                                All megakaryocytes showed strong positivity with the
Case History of Gray Platelet Syndrome                                          usual megakaryocytic markers. Additionally, the mega-
The bone marrow biopsy of a 36-year-old woman is shown                          karyocytes were strongly CD34-positive but alpha-gran-
who underwent a bone marrow biopsy (Fig. 2.21a–h) with                          ule marker (GF11B) was reduced (this marker was
the clinical supposed diagnosis of ITP. She and her children                    provided by the clinicians who excessively studied the
had episodes of life-threatening bleedings and the woman                        patients findings and reported this family). Erythropoiesis
and her children suffered from recurrent hematoma and pete-                     was slightly increased and left shifted, the granulopoiesis
chiae since childhood with platelets less than 45/nL. The                       appeared normal.
a b
GOM
Fig. 2.21 Gray platelet syndrome. (a) Routinely stained H&E section            megakaryocytes (CD42b). (f) Higher magnification of CD42b-positive
shows slightly hypercellular bone marrow. In this figure, rather small         megakaryocytes with hypolobulated nuclei. (g and h) All megakaryo-
megakaryocytes with hypolobulated nuclei can be seen. (b) Gomori’s             cytes are CD34-positive. Inset shows no or rather weak staining of
stain reveals a slight increase of diffusely dispersed reticulin fibers. (c)   megakaryocytes with an antibody to GF11B, whereas the erythropoi-
Erythropoietic cells are slightly increased and left shifted (CD71). (d)       etic cells are rather strongly positive
Normal appearing granulopoiesis (MPO). (e) Increase of rather small
2       Non-neoplastic Conditions with Quantitative and Qualitative Changes in Hematopoiesis       71
c d
CD71 MPO
e f
CD42b CD42b
g h
CD34
                a                                                           b
early changes
MGG MGG
 Fig. 2.22 Bone marrow biopsy of an 18-year-old male treated for          hematopoietic cells reveals diffusely dispersed large lymphoid imma-
 acute myelomonocytic leukemia at day 7. The bone marrow is aplastic      ture appearing cells (d, H&E). Immunohistochemical investigation
 showing edema and fibrinoid necrosis (a, MGG). Markedly dilated          shows CD10-positivity (e) and strong CD20-positivity (f) in these
 sinusoids are evident (b, MGG). Bone marrow biopsy of a 24-year-old      immature looking lymphoid cells uncovering these as hematogones. (g,
 male treated for acute T-cell leukemia at day 21. The bone marrow        h) Bone marrow biopsy of a 14-year-old girl treated for c-ALL at day
 shows areas with increased fatty tissue and areas with confluent hema-   21 showing normocellularity for this age (g, H&E) and the occurrence
 topoietic cells (c, H&E). High magnification of bone marrow field with   of small groups of megakaryocytes (h, CD42b)
 2                     Non-neoplastic Conditions with Quantitative and Qualitative Changes in Hematopoiesis       73
                        c                                                                d
intermediate changes
                        e                                                                f
intermediate changes
CD10 CD20
                        g                                                                h
late changes
CD42b
2.17.1 Caution                                                          very problematic in patients treated for acute B-cell leuke-
                                                                         mias. Sheets of promyelocytes might be confused with
In some patients, uncommon findings in regenerating bone                 acute promyelocytic leukemia. And finally, an increase of
marrows can be seen (Fig. 2.23). Such findings, which can                megakaryocytes (Fig. 2.23e, f), often combined with a dif-
be confused with a neoplastic process, comprise the appear-              fuse reticulin fibrosis can cause difficulties, especially in
ance of large sheets of promyelocytes (Fig. 2.23a–d), a                  patients with underlying myeloproliferative neoplasms.
marked increase in hematogones and a rather florid increase              Rarely, patients can show a marked delay in the regenera-
of megakaryocytes. An increase of hematogones can be                     tion of hematopoiesis.
a b
c d
Fig. 2.23 Uncommon findings in regeneration. This is the bone mar-       age (a, b: MGG). In the hypercellular areas nearly exclusively promy-
row of a 76-year-old woman treated for acute myelomonocytic leuke-       elocytic cells could be seen (c, d: H&E) The bone marrow biopsy of a
mia. The patient was pancytopenic after 3 weeks of treatment and         55-year-old male treated for AML (FAB M0) after 3 weeks therapy
received treatment with granuolcyte-stimulating factor. The bone mar-    shows a marked increase of megakaryocytes (e: H&E, f: CD42b)
row biopsy was partly highly hypocellular and partly hypercellular for
2       Non-neoplastic Conditions with Quantitative and Qualitative Changes in Hematopoiesis       75
e f
CD42b
2.18     Monoclonal Antibody Treatment                              tration, especially paratrabecular. After RTX therapy, an
          (Rituximab)                                                antigen loss of CD20 is found, and the infiltrates are mainly
                                                                     or completely composed of reactive T-cells (Fig. 2.24a–d). In
The chimeric murine-human anti-CD20 monoclonal anti-                 addition to an immunohistochemical stain with an antibody
body Rituximab (RTX) is in worldwide use for the treatment           to CD20, some more B-cell markers have to be used to avoid
of B-cell lymphomas, either alone or in combination with             the mistake of missing a residual neoplastic infiltrate.
standard regimens. There is a high response rate with a rather       Furthermore, areas of former lymphoma infiltration in para-
low toxic effect. The pathologists, investigating marrow             trabecular areas may reveal a reticulin fibrosis.
biopsies after RTX treatment, have to be familiar with the
morphologic and immunophenotypic sequelae this treatment
causes. The pitfalls for pathologists after RTX treatment            2.18.1 Caution
were first described by Douglas et al. in 1999 [39]. An
evaluation of different small B-cell lymphomas treated with         Since there is an antigen loss of CD20 in B-cells in patients
 RTX emphasizes the use of a panel of different antibodies to        treated with RTX, bone marrows have to be studied immuno-
 avoid false-positive and false-negative interpretations [40].       histochemically with additional B-cell markers, such as
 Especially, problematic are bone marrow biopsies of patients        CD19, CD22, and CD79a. If CD20 is the only B-cell marker
 with bone marrow infiltration by follicular lymphoma treated        used, neoplastic residual lymphoid cells might be missed. In
 with RTX. In these bone marrows, lymphocytic infiltrates            addition, also PCR studies for evaluation of clonality might
 can be detected in areas of former neoplastic lymphoid infil-       be necessary.
a b
c d
CD20 CD3
Fig. 2.24 RTX treatment. (a–d) Bone marrow biopsy of a 54-year-old   stained sections a slight fibrosis is recognizable. A nodular lymphocytic
male treated with RTX for follicular lymphoma. (a) (H&E) shows an    infiltrate (b, H&E) shows no reactivity with an antibody to CD20 (c),
area of former paratrabecular lymphoma infiltration. Even in H&E     the cells within this infiltrate are exclusively T-cells (d, CD3)
 2             Non-neoplastic Conditions with Quantitative and Qualitative Changes in Hematopoiesis                                                  77
               a                                                                 b
early 1 week
 Fig. 2.25 BM after TX. Bone marrow biopsy of a 47-year-old male               positive T-lymphocytes (e). Plasma cells (stained with an antibody to
 1 week after transplantation. Underlying disease was follicular lym-          Vs38c) are increased (f). Further studies with antibodies to light chains
 phoma. In H&E stained section, the bone marrow is highly hypocellular         kappa and lambda revealed polytypic plasma cells. Bone marrow
 (aplastic) (a). Higher magnification (b) reveals fat necrosis. (c–f) Bone     biopsy of a 12-year-old boy 11 months after bone marrow transplanta-
 marrow biopsy of a 29-year-old female 2 weeks after stem cell trans-          tion. Underlying disease was precursor T-cell neoplasia (T-lymphoblastic
 plantation. Underlying disease was AML. High magnification shows              leukemia). Avascular bone necrosis with empty osteocytic lacunae in
 dysplastic erythropoiesis and abnormal localization of immature               bone trabeculae is detected. Furthermore, a diffuse necrosis of marrow
 myeloid precursors (c, H&E). Immunohistochemically scattered small            cells can be seen (H&E, g, low magnification, h, high magnification)
 megakaryocytes (CD42b) are seen (d). There is an increase of CD3-
78                      2   Non-neoplastic Conditions with Quantitative and Qualitative Changes in Hematopoiesis
            c                                    d
1-2 weeks
                                                                                                       CD42b
            e                                    f
                                         CD3                                                            Vs38c
            g                                    h
12 month
39. Douglas VK, Gordon LI, Goolsby CL, White CA, Peterson              41. Orazi A, Weiss LM, Foucar K, Knowles DM. Chapter 47: Bone
    LC. Lymphoid aggregates in bone marrow mimic residual lym-             marrow morphologic changes after chemotherapy and stem cell
    phoma after rituximab therapy for non-Hodgkin lymphoma. Am J           transplantation. In: Knowles neoplastic hematopathology. 3rd ed.
    Clin Pathol. 1999;112:844.                                             Philadelphia: Wolters Kluwer/Lippincott, Williams and Wilkins;
40. Goteri G, Ranaldi R, Lucesole M, Filosa A, Capretti R, Pieramici       2014. p. 1165.
    T, Leoni P, Rubini C, Fabris G, LoMuzio L. Bone marrow his-        42. Lagoo AS, Gong JZ, Stenzel TT, Goodman BK, Buckley PJ, Chao
    topathological and molecular changes of small B cell lym-              NJ, Gasparetto C, Long GD, Rizzieri DA. Morphologic examina-
    phomas after rituximab therapy: comparison with clinical               tion of sequential bone marrow biopsies after nonmyeloablative
    response and patient’s outcome. Int J Immunopathol Pharmacol.          stem cell transplantation complements molecular studies of donor
    2006;19(2):421–31.                                                     engraftment. Arch Pathol Lab Med. 2006;130(10):1479–88.
                                 Bone Changes Found in Bone Marrow
                                 Biopsies                                                                                                  3
The skeleton function is diversified. It is the instrument for            Since it is impossible to show all these complex aspects,
direct locomotion and movement and protects vulnerable tis-            selected important entities will be discussed.
sues. It is important for the homeostasis of minerals, and it
shelters the bone marrow which provides the blood elements.
The metabolites of Vitamin D, parathyroid hormones, and                3.1        Osteodystrophy
calcitonin are among the most important regulators in the
complex mechanism for the control of the calcium mecha-                3.1.1      Renal Osteodystrophy
nism and the sound condition of bone. These complex mech-
anisms explain the vulnerability of trabecular bone which is           Renal osteodystrophy, a disease spectrum and not a uni-
variably affected in many primary or secondary diseases. A             form progressive bone disease [2], is caused by derange-
bone marrow biopsy can be helpful in diagnosis since mor-              ments in homeostatic and metabolic control of minerals.
phological changes can be detected before abnormalities are            This progressive bone disease histologically reveals a mix-
evident on X-rays. Bone remodeling and bone production                 ture of osteopenia, osteosclerosis, osteomalacia, and oste-
are typical features in pediatric marrows. However, even in            itis fibrosis cystica (Fig. 3.1a–d). Often reduction in
adults bone remodeling is continuing for mechanical support            hematopoiesis as well as fibrosis and edema give rise to
and to provide adequate calcium levels. This bone remodel-             the anemia of chronic renal disease. In any individual
ing in adults is morphologically not noticeable in the figura-         patient, different morphologic and clinic changes may pre-
tive sense of peculiar osteoblasts or osteoclasts [1]. The             dominate. Concerning the bone changes, these are affected
evaluation of trabecular bone on bone marrow biopsies com-             mainly by the manifestations of secondary hyperparathy-
prises an estimation of the thickness, size, and shape and a           roidism. Secondary hyperparathyroidism, in contrast to
general survey of the matrix and cellular components of                the primary form, shows various degrees of a malacic com-
bony trabeculae. Normally, a structural support for medul-             ponent. Distinctive forms of secondary hyperparathyroid-
lary cavities is evident by anastomosing bone spicules.                ism reveal fibro osteoclastic lesions. According to Delling
Continuously connected anastomosing bony trabeculae are a              [3], at least a rough estimation of renal osteodystrophy
pathologic finding. Careful evaluation of the mesenchymally            should be made. He suggests Type I: the main histologic
developed osteoblasts and osteocytes, and the osteoclasts,             finding is a fibro-osteoclastic morphology (Fig. 3.1a, b);
which are of hematopoietic origin, is essential. The mono-             Type II: marked osteomalacia is present (Fig. 3.1c, d);
nuclear osteoblasts are located in the periphery of trabeculae.        Type III: combination of Type I and II with low (a), moder-
In young patients, these cells often form a row along bony             ate (b), and marked (c) bone remodeling and “+” increased
trabeculae. Osteoclasts, mostly multinucleated cells, occupy           bone volume density and “−” decreased bone volume
small depressions on the bone’s surface, called Howship                density.
lacunae. A patchy distribution of osteoclasts along bony tra-
beculae is normal in young patients. Mineralized bone pre-             3.1.1.1 Caution
dominates and comprises osteocytes. Unmineralized newly                In patients with renal diseases, especially in dialysis patients,
formed bone matrix, the osteoid, is prominent in young chil-           a special stain for osteoid (non-mineralized bone) has to be
dren, but minimal in adults.                                           performed.
a b
c d
Fig. 3.1 Renal osteodystrophy. Bone marrow biopsy of a 59-year-old       46-year-old dialysis patient with marked osteomalacia (Ladewig’s
female dialysis patient with marked anemia. In the bone marrow biopsy,   stain). The blue bone is mineralized; the red color is indicative of oste-
marked fibro-osteoclastic lesions without a malacian component were      oid (non-mineralized bone)
seen (a: H&E; b: Ladewig’s stain). (c, d) Bone marrow biopsy of a
3       Bone Changes Found in Bone Marrow Biopsies                                                                                        83
a b
c d
Fig. 3.2 Hyperparathyroidism. (a–f) Bone marrow biopsy of a             seen. Excavated trabeculae with cystic formation (b, MGG), and marked
35-year-old woman diagnosed with primary hyperparathyroidism.           osteoclastic remodeling (c, d, MGG) is evident. (e, f) Paratrabecular
(a–d) In overview (a, MGG) distortion of trabecular bone structure is   coarse fibrosis extending into the marrow cavities (Gomori’s stain)
84                         3   Bone Changes Found in Bone Marrow Biopsies
e f
3.1.3        Osteopenia                                               resorption and formation throughout life. The aim is the
                                                                       adaption to stimuli and the preservation of structure and
Osteopenia is characterized by thinned bony trabeculae                 strength. The bone volume is regulated by balanced-coupled
(Fig. 3.3). As a generalized change, osteopenia is noted as            osseous remodeling in space within the skeleton and in time
osteoporosis; patchy osteopenia may occur in patients with             throughout life. The cortex is a low remodeling area, and the
renal disorders or in patients with neoplastic infiltrates, espe-      trabeculae are a high remodeling area. A complex interaction
cially myeloma patients. Osteoporosis is known to occur due            of pathologic stimulation of osteoclasts and inhibition of
to hormonal changes (menopause and andropause), meta-                  osteoblasts induces an imbalance in bone remodeling [6].
bolic dysfunctions (insulin deficiency), and due to adverse
effects of various chronic diseases affecting the skeleton:            3.1.3.1 Caution
these include cardiovascular, gastro-intestinal, musculo-             Osteopenic trabeculae are more common in females with an
skeletal, hematological, infectious, nephrological, oncologi-          increasing incidence in menopause. The age of the patient is
cal, rheumatic, and hepatic diseases. The skeletal integrity is        important in diagnosing osteopenia. If you find osteopenia,
maintained at remodeling sites by continuous cycles of                 check for comorbidities.
a b
c d
Fig. 3.3 Osteopenia. (a) bone marrow biopsy with marked osteopenia     trabeculae are markedly thinned (H&E). (e) Bone marrow biopsy of a
in a 41-year-old woman after bone marrow transplantation due to fol-   25-year-old male after bone marrow transplantation because of acute
licular lymphoma. Note the filigree disconnected bony trabeculae       myeloid leukemia. The trabeculae appear osteopenic; they are sur-
(H&E). (b) Bone marrow of a 51-year-old female with liver cirrhosis.   rounded by adipose tissue. (H&E). (f) Bone marrow biopsy of a
Trabeculae are disconnected and markedly thinned (H&E). (c) Bone       22-year-old male who underwent transplantation because of acute
marrow biopsy of a 42-year-old male patient who had acute myeloid      myeloid leukemia. Bone marrow is normocellular, but the trabeculae
leukemia and chemotherapy (H&E). (d) Bone marrow biopsy of a           are thinned for this age (H&E)
31-year-old female with pure red cell anemia and thymoma. The bony
86                         3   Bone Changes Found in Bone Marrow Biopsies
e f
a b
Fig. 3.4 (a, b) Bone marrow biopsy of a 73-year-old male with chronic intestinal disease (colitis ulcerosa). Ladewig’s stain highlights the
increased osteoid (red)
88                                                                                        3   Bone Changes Found in Bone Marrow Biopsies
3.1.5     Osteopetrosis (Marble Bone Disease)                          encoding the a3 subunit of the vascular proton pump, which
                                                                        mediates acidification of the bone osteoclast interface. This
In osteopetrosis (OP), a heterogeneous disease group with               defect could be located to the osteoclasts [8]. This disorder is
genetic disorders, excessive unbalanced bone formation is evi-          often lethal within the first years of life and stem cell trans-
dent leading to trabecular sclerosis (Fig. 3.5). The result is a        plantation remains the only curative therapy option. An adult
diminution of the marrow spaces required for an effective               form of OP (called benign osteopetrosis) is diagnosed in late
hematopoiesis. The disease can cause pancytopenia, extra-               adolescence or adulthood and presents as anemia.
medullary hematopoiesis resulting to hepatosplenomegaly,
cranial nerves compression, and severe growth failure. The              3.1.5.1 Caution
underlying pathophysiological defect in all types of OP is the          Keep in mind, that the cause of hepatosplenomegaly in child-
failure of the osteoclasts to reabsorb bone, leading to thick-          hood could be osteopetrosis. Also remember that OP can
ened sclerotic bone with poor mechanical properties. Recently,          occur in young adults who undergo a bone marrow biopsy
mutations have been identified in the ATP 6 (ICIR GI gene)              because of anemia.
a b
GOM GOM
c d
Lad Lad
Fig. 3.5 (a–d) Bone marrow biopsy of a 4-year-old girl diagnosed with osteopetrosis. There is a jumbled bone formation causing obliteration of
the marrow cavities (a and b Gomori’s stain, c and d are stained according to Ladewig)
3       Bone Changes Found in Bone Marrow Biopsies                                                                                        89
3.1.6        Paget’s Disease of Bone                                   involvement of pelvis, femur, scull, clavicles, ribs, and tibia
                                                                        is found. The osteoclasts are enlarged and show a variable
Paget’s disease of bone is the second most common meta-                 number of nuclei, some of the osteoclasts revealing more
bolic bone disease in the Western world [9]. Paget’s disease            than 100 nuclei. Due to the rapid bone formation in many
is a localized disorder of uncontrolled bone remodeling.                cases, a typical mosaic pattern of the trabecular bone is
Osteoclastic hyperactivity stimulates osteoblasts resulting             seen.
in an overgrowth of bone formation (Fig. 3.6). This osteo-
clastic hyperactivity followed by compensatory osteoblas-               3.1.6.1 Caution
tic activity leads to a structurally disorganized mosaic of             In the early stage of Paget’s disease, the morphology may
woven bone, which is less compact and more susceptible to               mimic the changes seen in primary hyperparathyroidism.
deformities and fracture than normal adult lamellar bone.                 Osteoclasts in Paget’s disease are markedly increased in
 The main symptom is bone pain. In most cases, a focal                  number and size and can contain up to 100 nuclei.
a b
Fig. 3.6 Paget disease. (a, b) The trabecular bone shows the characteristic mosaic pattern. Huge osteoclasts cover the trabecular surface (a:
H&E; b: MGG). (c) Typical giant osteoclast with multiple nuclei (H&E)
90                                                                                        3   Bone Changes Found in Bone Marrow Biopsies
In patients with known or suspected non-hematological                  mens from different areas of a single iliac crest are recom-
malignancies, the indications for a bone marrow biopsy                 mended [1]. In adults, the most common primary tumors
include the following:                                                 metastasizing into the bone marrow are breast-, lung-, prostate-,
                                                                       gastrointestinal tract-, renal cell, and genitourinary carcino-
1.   Integral part of the initial staging                              mas, but also metastases from malignant melanomas and
2.   Metastases are suspected clinically                               rarely sarcomas can be detected.
3.   For monitoring the therapy and follow-up                             In children, the most common tumors, metastasizing
4.   For unexplained fever                                             into the bone marrow are neuroblastomas, Ewing sarco-
5.   For hypercalcemia                                                 mas, rhabdomyosarcomas, osteosarcomas, and rarely
6.   For high LDH-levels                                               medulloblastomas.
7.   For suspicious X-ray or scan                                         Marrow infiltration by metastatic tumor in most cases is
                                                                       focal, but diffuse infiltration can occur. In the bone marrow,
    A bone marrow biopsy is useful in staging and determin-            areas with metastases commonly reveal reticulin and/or col-
ing of bone marrow metastases of solid tumors. The diagno-             lagen fibrosis and are most pronounced in marrows with
sis of bone marrow metastases is important because of                  greater degrees of marrow infiltration. Other stromal reac-
treatment methods and also survival. Anemia, thrombocyto-              tions may include edema, increased blood vessel formation,
penia, leukopenia, bi-cytopenia, and pancytopenia, but also            an inflammatory infiltrate at the margin of the neoplasm and
leukemoid reactions and leucoerythroblastic reactions can be           lytic, as well as osteoblastic trabecular changes. Lytic
found after the metastatic infiltration of the bone marrow.            changes show an overall loss of bony trabeculae with often
Occasionally, bone marrow metastases are verified without              an increased number of osteoclasts. Osteoblastic changes
any changes in hematologic parameters. Metastases in the               are combined with new bone formation and an increased
bone marrow can be seen in patients with widely metastatic             number of osteoblasts, occasionally revealing a pagetoid
solid tumors, but also in tumors firstly diagnosed by bone             appearance.
marrow biopsies.                                                          In many, or even most cases, a classification of the metas-
    The incidence of bone marrow metastases by various                 tases in the bone marrow is possible with immunohistochem-
malignant tumors has changed over the years due to the ear-            ical stains (Figs. 4.1, 4.2, 4.3, and 4.4). Overall, the
lier diagnosis of some tumors and due to the improved detec-           immunohistochemical characterization of metastatic carci-
tion of micrometastases in the bone marrow. Since metastatic           nomas to the bone marrow shows a good correlation with the
infiltrates are usually focal, the length of the bone marrow           established staining pattern of the primary tumors [2].
biopsy material is important for the detection of metastases,             Some examples of neoplastic bone marrow infiltrations
and bilateral bone marrow biopsies or double biopsy speci-             (Figs. 4.1–4.9) are shown.
a b
c d
CKAE1 CK7
e f
GATA3 CD71
g h
estrogen progesteron
Fig. 4.1 (a–h) Case 1 (breast carcinoma): Bone marrow biopsy of a             The tumor cells are stained with an antibody to CKAE1/3 (c) and with
69-year-old female with suspected carcinoma. The patient presented            an antibody to CK7 (d). GATA3-positivity (e) gives the link to a metas-
with bone pain, anemia, and increased LDH. In H&E stained sections            tasis from a breast carcinoma. A strong positivity for all tumor cells is
(a, b), an infiltration by carcinoma is seen. Hematopoiesis is replaced       seen with an antibody to CD71 (Transferrin receptor), (f). The tumor
by an extensive neoplastic epithelial tumor with new bone formation.          cells are estrogen-positive (g) and progesterone-negative (h)
94                                                                                 4   Non-hematological Malignancies in the Bone Marrow
a b
c d
CKAE1 CK7
e f
CK20 TTF1
Fig. 4.2 (a–f) Case 2 (lung carcinoma): A 34-year-old female devel-     H&E stained sections, the bone marrow shows extensive replacement
oped dyspnea after giving birth by Caesarean section. She developed     by metastatic adenocarcinoma with marked new bone formation (a, b).
pleural effusion. Pneumonia was suspected, but despite treatment, her   The tumor cells are strongly cytokeratin-positive (c, CKAE1/3) and
condition worsened. MR-tomography revealed pathologic signals in the    show positivity for CK7 (d) and also CK20 (e). TTF-1-positivity of the
whole spinal column, and a bone marrow biopsy was performed. In         tumor cells (f) is suggestive of a primary in the lung
4       Non-hematological Malignancies in the Bone Marrow                                                                                      95
a b
c d
CKAE1 CD56
e f
synaptophysin TTF1
Fig. 4.3 (a–h) Case 3 (small cell carcinoma of lung): A 55-year-old      patchy infiltration by rather small tumor cells (a, b: H&E). Strong reac-
female presented with leucocytosis and polyglobulia. Medical exami-      tivity of the tumor cells is seen with antibodies to CKAE1/3 and CD56
nations were susceptive of a centrally located carcinoma of the bron-    (c, d). Few tumor cells are weakly synaptophysin-positive (e). Strong
chus. A bone marrow biopsy was performed revealing a dense               reactivity of the tumor cells with an antibody to TTF1 confirming the
infiltration by small carcinoma cells with neuroendocrine differentia-   primary tumor in the lung (f)
tion, confirming small cell carcinoma of the bronchus. Diffuse and
96                                                                               4   Non-hematological Malignancies in the Bone Marrow
a b
c d
CK5/6
Fig. 4.4 (a–d) Case 4 (squamous cell carcinoma): A 59-year-old male    The bone marrow was infiltrated by metastases of squamous cell carci-
with squamous cell carcinoma of the hypopharynx, diagnosed             noma (a–c: H&E). Note the new bone formation (a, b). Squamous cell
12 months previously, developed increasing pancytopenia. A bone mar-   carcinoma was immunohistochemically verified by staining with an
row biopsy was done with the suspected diagnosis of myelodysplasia.    antibody to CK5/6 (d)
4   Non-hematological Malignancies in the Bone Marrow                                                                                               97
a b
c d
                                                                S100                                                                    Langerin
e                                                                          f
CD1a CD1a
Fig. 4.5 (a–f) Case 5 (Langerhans cell histiocytosis): A 78-year-old           hypercellular (a, H&E) with focal nests of atypical histiocyte-like cell
icteric patient was admitted to the hospital with itching and exanthema.       infiltrates (b, H&E). These atypical cells were stained with antibodies
A skin biopsy was performed and diagnosed as Langerhans cell histio-           to S-100-protein (c), Langerin (d) and CD1a (e, f) revealing marrow
cytosis. Because of increasing bone pain and pancytopenia a bone mar-          involvement by Langerhans cell histiocytosis
row biopsy was performed. The bone marrow was moderately
98                                                                                4   Non-hematological Malignancies in the Bone Marrow
a b
c d
CD21 CD23
e f
CD106 CD35
Fig. 4.6 (a–f) Case 6 (FDC sarcoma): A 71-year-old male patient was     nuclear chromatin, distinct nucleoli and a delicate nuclear membrane
diagnosed with follicular dendritic cell sarcoma infiltrating urinary   (a, b, H&E). The tumor cells were strongly CD21-positive (c), but neg-
bladder, liver, and bone marrow. The bone marrow biopsy revealed a      ative with an antibody to CD23 (d). Furthermore, the tumor cells were
patchy paratrabecular infiltrate of large tumor cells with vesicular    stained with antibodies to CD106 (e) and CD35 (f)
4       Non-hematological Malignancies in the Bone Marrow                                                                                 99
a b
c d
NBL f 17 Chrom
e f
Synapt MGG
Fig. 4.7 (a–f) Case 7 (neuroblastoma): At the age of 11 years, a girl   Multifocal dense to patchy infiltration by neuroblastoma is seen. The
was diagnosed with neuroblastoma originating from the left adrenal      tumor cells reveal abundant granular eosinophilic cytoplasm resem-
gland with widespread metastases. The diagnosis was confirmed by        bling ganglion cells (a, b H&E). The tumor cells are stained with a
immunohistochemistry and nMYC gene amplification. The bone mar-         neuroblastoma marker (c), chromogranin (d), and synaptophysin (e). In
row was also infiltrated indicating an advanced stage of disease.       some areas, ganglioneuroblastoma can be seen (f, MGG)
100                                                                               4   Non-hematological Malignancies in the Bone Marrow
a b
c d
NSE CD56
Fig. 4.8 (a–d) Case 8 (neuroblastoma): In this case of a 2-year-old     tumor (a, H&E). The infiltrating cells are immunohistochemically reac-
boy, the rather fragmented bone marrow biopsy is infiltrated by tumor   tive with antibodies to a neuroblastoma marker (b), NSE (c), and CD56
cells of neuroblastoma with the morphology of a small blue round cell   (d)
4       Non-hematological Malignancies in the Bone Marrow                                                                                    101
a b
c d
FVIII-rag
Fig. 4.9 (a–d): Case 9 (angiosarcoma): A 75-year-old woman devel-         area). Higher magnification reveals irregular dissecting vascular chan-
oped angiosarcoma of the breast 32 months after lumpectomy and            nels lined by pleomorphic cells with prominent nucleoli (b, c, H&E).
radiotherapy for a ductal breast carcinoma. The angiosarcoma infil-       Reactivity with an antibody to FVIII-rag (d) in the tumor cells and in
trated the chest wall, liver, and bone marrow. In the bone marrow, a      megakaryocytes (top right)
patchy infiltrate absorbing a whole marrow space is seen (a, H&E, right
102                                                                               4   Non-hematological Malignancies in the Bone Marrow
a b
MGG Vs38c
c d
kappa lambda
Fig. 5.1 Reactive plasmacytosis. (a) In the hypercellular bone marrow,   d) With antibodies to light chains (c: kappa; d: lambda) the plasma cells
a marked increase of mature plasma cells is seen (MGG). (b) The          are polyclonal
plasma cells are highlighted with a plasma cell marker (Vs38c). (c,
5   Increase of Plasma Cells, Reactive and Neoplastic                                                                        105
a b
c d
e f
CD56 κ λ
Fig. 5.2 This shows morphologic variants of plasma cells in plasma         roglobulinemia. (c) The bone marrow of this 75-year-old woman is
cell myelomas. (a, b) Bone marrow infiltration by plasma cell myeloma      infiltrated by plasma cell myeloma with foamy appearance of the
in a 53-year-old woman: on the one hand (a left side), the patchy infil-   plasma cells. (d) In higher magnification, some multinucleated cells
trating plasma cells show a characteristic plasmacytic appearance, on      with prominent eosinophilic nucleoli are visible. (e) The neoplastic
the other hand (right side) the tumor cells reveal an abundant eosino-     plasma cells are reactive with an antibody to CD56. (f) Clonality is
philic morphology (H&E). (b) Higher magnification of the plasma cells      evident by expression of the light chain kappa (left), whereas the plasma
with eosinophilic cytoplasm. These plasma cells are called “flame          cells are lambda-negative (right)
cells.” Flame cells can occasionally be observed in Waldenström’s mac-
5       Increase of Plasma Cells, Reactive and Neoplastic                                                                                        107
a b
c d
Vs38c Vs38c
e f
κ λ
Fig. 5.3 Case 1. This shows dense bone marrow infiltration of plasma        remodeling is also highlighted with the antibody Vs38c, which is
cell myeloma in a 39-year-old woman with plasmacytic appearance and         known to stain osteoblasts. (e) The plasma cells are not reactive with an
abnormal expression of CD23. (a, b) Dense infiltration of the bone mar-     antibody to kappa light chain. (f) Monoclonality of the plasma cell
row by plasmacytic type of plasma cell myeloma (H&E). (c) Strong            population is shown by strong lambda-positivity. (g) The monoclonal
staining of plasma cells with an antibody to Vs38c (antigen p63: this       plasma cells are reactive with an antibody to CD23. (h) Only single
protein is an intracellular type II transmembrane protein which is local-   plasma cells are Cyclin D1-positive
ized in the rough endoplasmic reticulum) (d) The increased osseous
108                                                                                        5   Increase of Plasma Cells, Reactive and Neoplastic
g h
CD23 Cyclin D1
a b
c d
CD138 λ
Fig. 5.4 Case 2. This shows dense bone marrow infiltration by an ana-      cells shows polymorphic blasts with clear irregularly formed nuclei and
plastic variant of plasma cell myeloma. In H&E stained sections, the       multiple nucleoli (H&E). (c) Strong reactivity of the tumor cells with a
morphology of the infiltrating cells does not reveal the typical plasma-   plasma cell marker (CD138). (d) The neoplastic plasma cells are
cytic differentiation. (a) Highly hypercellular bone marrow with dense     lambda-positive (not shown is kappa-negativity)
infiltration of tumor cells (H&E). (b) High magnification of the tumor
5       Increase of Plasma Cells, Reactive and Neoplastic                                                                                       109
a b
c d
CD56 CD56
e f
GOM Vs38c
Fig. 5.5 Case 3. Bone marrow biopsy of a 63-year-old female with           CD56-positivity of plasma cells, also paratrabecular endothelial cells
dense sclerosis (sclerosing type of plasma cell myeloma). (a) Even in      show reactivity with CD56. (e) Gomori’s stain reveals collagen fibrosis.
H&E stained section a dense sclerosis can be seen. In addition, there is   (f) Positivity of the plasma cell with Vs38c. (g) The infiltrating plasma
marked osseous remodeling with irregularly formed bone. (b) Higher         cells are monoclonal expressing the light chain kappa. (h) No reactivity
magnification shows plasma cell infiltration within the sclerotic stroma   of the plasma cells with an antibody to light chain lambda
(H&E). (c) The plasma cells are CD56-positive. (d) In addition to the
110                                                                           5   Increase of Plasma Cells, Reactive and Neoplastic
g h
κ λ
a b
CD117c
Fig. 5.6 Plasma cell myeloma with expression of CD117c (a H&E, b            vessel wall and small cloud-like amyloid deposits in the interstitium
CD117c). (c–h) Plasma cell myeloma with amyloidosis. In a 75-year-          (left from vessel wall). (f) Amyloid deposits within a vessel wall in a
old male patient with plasma cell myeloma, amyloid deposits are easily      rather large vessel in the subcortical area. (g, h) In the bone marrow of
visible in Giemsa stained sections. (c) Even at low magnification, thick-   a 67-year-old woman with treated plasma cell myeloma extensive inter-
ened vessel walls with bluish deposits can be seen. (d) Plasma cells are    stitial amyloid deposits are evident (g H&E) (h) immunohistochemical
surrounding a small vessel with amyloid deposits. (e) Amyloid within a      stain with an antibody to amyloid P
112                        5   Increase of Plasma Cells, Reactive and Neoplastic
c d
e f
g h
Amyloid P
The investigation of the bone marrow by bone marrow 4. Chronic inflammation, “fibrotic” type: There is an
biopsy is one of the most valuable diagnostic tools for evalu-       increase in reticulin and/or collagen fibers with reduction
ating hematologic disorders. Furthermore, bone marrow                of fat cells and hematopoiesis (“sclerosing myelitis”). In
analysis has great importance in the evaluation of non-             most cases, reactive T-lymphocytes and plasma cells are
hematologic conditions. Various chemical toxins, including           increased, edema might be present and even new bone
minerals, as well as radiation may severely affect the bone          formation can be evident. This condition is often found in
marrow cells and stroma. In patients with fever of unknown           infections (especially HIV) and lupus as well as other
origin (FUO) and in patients with autoimmune deficiency              autoimmune diseases [2] (see Fig. 6.3a, b).
syndrome (AIDS), the marrow may reveal various microor- 5. Chronic inflammation “proliferative” type: The
ganisms, such as leishmaniasis and tuberculosis, and fungal          bone marrow is either normocellular or hypercellular
as well as viral infections.                                         with an increase of reactive T-lymphocytes, plasma
    Roughly, nonspecific inflammatory changes in the bone            cells, mast cells, and/or eosinophils. This condition is
marrow can be subdivided into several types; however, these          mainly seen in patients with rheumatoid arthritis [3]
inflammatory changes may occur in combined forms.                    (see Fig. 6.3c, d).
                                                                  6. Chronic inflammation, “leukemoid” type: The bone
1. Acute inflammation, “exudative” or “necrotic” type:               marrow is slightly or marked hypercellular with increased
    Necrosis of hematopoietic cells and capillaries as well as       neutrophils or eosinophils and often increased megakary-
    edemas may be present (see Fig. 6.1a, b). Some hemato-           ocytes. There is an increase in the estimated M:E ratio,
    poiesis may be left. In addition, bone necrosis, especially      but maturation synchrony is often unaffected. Due to
    caused by chemotherapy, might be present.                        increases in granulopoietic cells, quantitative bone mar-
2. Acute inflammation, “hemorrhagic” type: Extensive                 row differential cell counts reveal an increased M:E ratio
    hemorrhage is associated with hypoplasia of hematopoi-           [4] (see Fig. 6.3e, f).
    etic cells. This condition is seen, for example, in hepatitis 7. Chronic inflammation “granulomatous” type (see
    B infection or in patients with severe histiocytic activa-       Fig. 6.4a–f): In this condition, giant cell granulomas,
    tion syndrome (see Fig. 6.1c, d) [1].                            lipoid granulomas, or epithelioid cell granulomas are
3. Chronic inflammation, “atrophic” type: In addition to             seen. Granulomas are nodular aggregates of inflamma-
    edema, an increase of lymphocytes (mainly reactive               tory cells, mainly consisting of epithelioid cells (Fig. 6.4a,
    T-cells), plasma cells, and mast cells is often seen.            b) and/or giant cells with various numbers of lympho-
    Hematopoiesis is markedly reduced. Synonymously to               cytes, plasma cells, mast cells, and fibroblasts in and
    “atrophic” type, this condition is known as gelatinous           around the granulomas. Occasionally, inclusion bodies
    transformation, serous atrophy, or exudative myelitis.           within giant cells can be found. Granulomas can be seen
    Such changes may occupy all marrow spaces or circum-             in hypo-, normo-, or hypercellular bone marrow as a
    scribed marrow areas. This condition is seen in various          response to various processes including sarcoidosis,
    diseases, especially in patients with poor nutrition or mal-     tuberculosis, collagen diseases, chemotherapy, fungi,
    nutrition, chronic infections, and malignancies. The bone        drugs, and various malignant diseases (paraneoplastic),
    marrow is markedly hypocellular; also the fat cells are          especially lymphomas [5]. Granulomas can also be found
    reduced (see Fig. 6.2a–h).                                       without any manifest diseases [6].
a b
c d
Fig. 6.1 Acute inflammation. (a, b) Bone marrow of a 16-year-old          edematous stroma (H&E). (c, d) Hemorrhagic inflammation of the
pancytopenic male after chemotherapy because of a Ewing sarcoma.          bone marrow in a pancytopenic 55-year-old female with hepatitis B
There is exudative inflammation with reduced hematopoiesis and            (H&E)
a b
MGG MGG
Fig. 6.2 Chronic inflammation, atrophic type. (a, b) Gelatinous trans-    bone marrow with gelatinous transformation. (f) Another bone marrow
formation of all marrow spaces in an 18-year-old pancytopenic female      space of the same patient stained with PAS. In this marrow space, a few
with anorexia nervosa. Hematopoiesis is highly reduced; marked exu-       hematopoietic cells are evident. (g) Immunohistochemical stain with an
dative myelitis is found (MGG). (c, d) In H&E stained section, the mar-   antibody to CD71 shows a few erythropoietic cells. (h) A single mega-
row spaces in this young woman reveal stromal gelatinous                  karyocyte is detected using an antibody to CD42b
transformation without hematopoietic cells. (e) Gomori’s stain of the
6       Inflammatory and Infectious Diseases                  117
c d
e f
GOM
g h
CD71 CD42b
a b
c d
CD3
e f
MPO
Fig. 6.3 Chronic inflammation. (a, b) Fibrotic type of inflammation in        lar (c, MGG). Immunohistochemical stain with an antibody to CD3
a 26-year-old female patient with systemic lupus. The bone marrow             shows increased T-lymphocytes (d). (e, f) Leukemoid type of chronic
biopsy was taken because of marked anemia and slight leukopenia as            inflammation in a 67-year-old male patient with colonic carcinoma.
well as thrombocytopenia. In H&E stained section, the marrow is hypo-         Overview of the MGG stained bone marrow shows highly hypercellular
cellular for age (a), Gomori’s stain reveals irregular fibrosis (b). (c, d)   marrow spaces (e), mainly caused by an increased granulocytopoiesis
Proliferative type of inflammation in a 56-year-old woman with a long         (f), visualized by immunohistochemical stain with an antibody to MPO
history of rheumatoid arthritis. The bone marrow is highly hypercellu-
6       Inflammatory and Infectious Diseases                                                                                                 119
a b
c d
e f
Fig. 6.4 Chronic inflammation (a, b) Chronic inflammation, granulo-      thelioid cell granulomas. This patient suffered of night sweats and dys-
matous type. H&E stained bone marrow in a young 28-year-old woman        pnea as well as back pain. PCR studies of the bone marrow with this
with fever of unknown origin. An epithelioid cell granuloma is closely   marked granulomatous infiltrates for germ analysis, including myco-
attached to the bone. Clinical workup revealed sarcoidosis. (c–f) The    bacteria were negative. Clinical work-up finally yielded sarcoidosis
bone marrow biopsy of this 52-year-old woman reveals masses of epi-      (H&E)
120                                                                                                6   Inflammatory and Infectious Diseases
6.1       Infectious Diseases                                           lation. Patients with XLP are usually young male children
                                                                         with mutations of the SAP/SH2D1A gene. If these patients
For most systemic infections, the bone marrow reveals non-               survive the EBV infection (infectious mononucleosis), they
specific changes which, however, are compatible with a reac-             might develop lymphomas or dysgammaglobulinemia.
tive, non-neoplastic process. Such reactive changes might be
the occurrence of granulomas or hemophagocytosis (see                    Case History 1
Figs. 6.5 and 6.6). In the following chapter, various charac-            A 68-year old female presented to Intensive Care Unit with
teristic morphological features are described.                           high fever, weight loss, and rash as well as pancytopenia.
                                                                         Furthermore, evidence of hemophagocytosis with hyperferri-
                                                                         tinemia was found. Pneumonia and hepatitis B were diagnosed.
6.1.1     Hemophagocytic Syndrome                                       She had a past history of multidrug-resistant tuberculosis.
                                                                            In the hypercellular bone marrow biopsy marked hemo-
Hemophagocytic syndromes (HPS) are either primary/                       phagocytosis was found, suggestive of secondary infection-
familiar diseases or secondary reactive disorders caused by              associated hemophagocytic syndrome (Fig. 6.5a, b) [9].
various infections, neoplasms, or vascular collagen diseases             Despite intensive treatment, the patient died of multiple
[7]. The bone marrow is hypercellular with myeloid hyper-                organ failure.
plasia in many cases (Fig. 6.5), but also hypocellularity can
be found. The bone marrow (see Fig. 6.5) shows an increase               Case History 2
of histiocytes with phagocytosis of erythropoietic and gran-             A 87-year-old male patient was admitted with massive epi-
ulopoietic cells and occasionally also platelets. While hemo-            staxis and high fever. Past history revealed cardiac problems
phagocytosis is easily detected, the underlying disease                  and carcinoma of the urinary bladder 12 years previously. At
causing hemophagocytosis often remains obscure. The                      admission, the patient was pancytopenic: Leuko 0.93·109/L,
infection-associated HPS can be caused by parasitic or bac-              Ery 4.24·1012/L, Hb 11.8 g/dL, Hkt 36.9%, MCV 87.0 fL,
terial infections, but in most cases HPS is caused by viral              MCH 27.8 pg, MCHC 32.0 g/dL, Thrombo 51·109/L, Ferritin
infections, especially EBV. EBV-associated HPS compro-                   was increased with 888.63 ng/mL. Bone marrow cytology
mises two distinct entities: (1) patients with sporadic hemo-            revealed hypocellularity with dysplastic erythropoietic cells
phagocytic lymphohistiocytosis and (2) patients with                     and hemophagocytosis (Fig. 6.5c, d) A bone marrow biopsy
X-linked lymphoproliferative disorder (XLP) (Fig. 6.6).                  showed highly hypocellular bone marrow with increased
Both diseases originate from an abnormal T-cell immune                   macrophages, many of which showed hemophagocytosis,
response to the cells infected by EBV. XLP is a rare primary             especially erythrophagocytosis (Fig. 6.5e–h). The patient
immunodeficiency first described in 1975 by Purtilo et al.               was treated symptomatically and died 2 days after
[8]. The disease is accompanied by severe immune dysregu-                admission.
a b
CD68
Fig. 6.5 (a) (Case history 1) Hypercellular hemorrhagic bone marrow      sive erythrophagocytosis. Highly hypocellular bone marrow (H&E e)
(H&E) (b) (Case history 1) Immunohistochemically, using an antibody      with an increase of macrophages with erythrophagocytosis (H&E f). (g,
to CD68, many macrophages with hemophagocytosis are present. (c–h)       h) Immunohistochemical staining with an antibody to CD163 high-
(Case history 2) Bone marrow smear with dysplastic erythropoietic cell   lighting the macrophages showing erythrophagocytosis
and macrophage with erythrophagocytosis. (d) Macrophage with mas-
6       Inflammatory and Infectious Diseases                  121
c d
e f
g h
CD163 CD163
Case History 3: X-linked Lymphoproliferative                          tions of the SAP/SH2D1A gene. A bone marrow biopsy was
Syndrome (XLP) (Fig. 6.6)                                             performed (Fig. 6.6). The bone marrow was hemorrhagic
A 3-year-old male child was admitted from another hospital            with decreased hematopoiesis, increased T-lymphocytes, and
with sustained high fever non-responsive to antibiotic ther-          occurrence of B-blasts. Hemophagocytosis was evident and
apy. The child was healthy until now, family history was unre-        EBV infection could be visualized (Fig. 6.6a–h). Despite
markable. The child was transferred with the supposed                 immediate intensive treatment, the child died soon after
diagnosis of EBV infection. Clinical workup revealed muta-            admission.
a b
CD71
c d
MPO CD61
Fig. 6.6 Infectious hemophagocytosis. (a) H&E stained bone marrow     CD61. (e) T-lymphocytes are markedly increased (CD3). (f) There are
of the 3-year-old boy shows hypocellular hemorrhagic marrow spaces.   many activated looking B-blasts (CD20). (g) Staining with a macro-
(b) Decreased erythropoietic cells stained with an antibody           phage marker (CD163) shows many macrophages with hemophagocy-
to CD71. (c) Granulopoietic cells are highly decreased (MPO).         tosis. (h) EBV infected cells visualized by EBER
(d) There are still many megakaryocytes stained with an antibody to
6       Inflammatory and Infectious Diseases                                                                       123
e f
CD3 CD20
g h
CD163
6.2       Visceral Leishmaniasis (Fig. 6.7)                            Kala-azar, black disease or black fever) is essential for the
                                                                        patient’s survival, especially when this infection appears
The recognition of morphological changes in the bone mar-               clinically unsuspected in non-endemic areas. Visceral leish-
row in cases with visceral leishmaniasis (also known as                 maniasis is caused by the protozoan parasite Leishmania
a b
c d
e f
Fig. 6.7 Infectious leishmaniasis. (a, b) Moderately hypercellular      within macrophages (inset), occasionally outside of nucleated cells.
bone marrow (overview a) with increased hematopoiesis. A slightly       (e) Amastigotes in MGG stained section (×1000). (f) Iron stained sec-
higher magnification shows inclusions in macrophages (b). (c, d) high   tion with iron-negative parasites (×1000)
magnification (H&E ×1000) reveals masses of amastigotes mainly
6   Inflammatory and Infectious Diseases                                                                                   125
Donovani and is transmitted into the patients system by the      extensive clinical investigations could not detect the reason,
bite of infected sandflies. More than 90% of the world’s         and the patients conditioned worsened dramatically, a bone
cases are reported from the Indian subcontinent [10]. Many       marrow biopsy was performed. In the bone marrow, a mas-
different Leishmania species have been identified, in            sive Leishmania Donovani invasion was found (Fig. 6.7a–f),
Mediterranean areas Leishmania infantum is the commonest         confirmed by PCR investigation at an Institute for tropical
form of disseminated visceral infections.                        medicine. The patient received specific treatment and recov-
   The clinical presentations of patients infected with leish-   ered. Anamnesis of this patient revealed a holiday in the
maniasis are in most cases fatigue, hepatosplenomegaly,          South of Spain 5 months previously; otherwise, this Austrian
unexplained fever as well as pancytopenia.                       patient had not been in a foreign country, where he could
   In the bone marrow biopsy numerous histiocytes, occa-         have been infected.
sionally lymphohistiocytic aggregates with mainly intracel-         The bone marrow is hypercellular for age. Numerous
lular and occasionally extracellular protozoa can be seen (see   amastigotes in macrophages, occasionally in the stroma,
Fig. 6.7a–f). Aggregates of this Leishmania Donovani bodies      were seen and the diagnosis of visceral leishmaniasis was
may be seen in the form of a ring shape or flower shape,         made.
mimicking fungal infection.
a b
c d
Fig. 6.8 Parvovirus infection. (a) The bone marrow of the 25-year-old    for megakaryocytes (CD42b) no such cells can be found (there are only
female is moderately hypocellular for age (H&E). (b) High magnifica-     some aggregates of thrombocytes in the lower right corner). (h)
tion of an erythron reveals extremely large pronormoblasts. (c, d) In    Granulopoiesis (stained with an antibody to MPO) is moderately
some erythropoietic cells, eosinophilic nuclear inclusions can be seen   decreased
H&E. (e, f) Appearance of “clear” nuclei (H&E). (g) Using an antibody
6       Inflammatory and Infectious Diseases                                                                               127
e f
g h
CD42b MPO
a b
c d
Fig. 6.9 Mycobacterium tuberculosis infection. (a) The bone marrow        Langhans type detected with an antibody to transferrin receptor
is moderately hypercellular for age (H&E). (b, c) Closely attached to a   (CD71). (g) Rather weak staining of epithelioid cells and histiocytes
trabecula a single caseating granuloma is found (H&E). (d) PAS-stain      with an antibody to CD14. (h) Iron deposits (Berlin blue) are surround-
was performed to exclude fungal disease. (e) Reticulin fibers within      ing the granuloma
the granuloma (Gomori’s stain). (f) A multinucleated giant cell of
6       Inflammatory and Infectious Diseases                129
e f
GOM CD71
g h
CD14 BBL
References                                                                 7. Chen CJ, Huang YC, Jaing TH, Hung IJ, Yang CP, Chang LY, Lin
                                                                              TY, et al. Hemophagocytic syndrome: a review of 18 pediatric
                                                                              cases. J Microbiol Immunol Infect. 2004;37:157–63.
 1. Jha A, Adhikari RC, Sarda R. Review article. Bone marrow               8. Purtilo DT, Cassel CK, Yang JP, Harper R. X-linked recessive pro-
    evaluation in patients with fever of unknown origin. J Pathol.            gressive combined variable immunodeficiency (Duncan’s disease).
    2012;2:231–40.                                                            Lancet. 1975;1(7913):935–40.
 2. Hasserjian RP. Reactive versus neoplastic bone marrow. Problems        9. Fisman DN. Hemophagocytic syndromes and infection. Emerg
    and pitfalls. Arch Pathol Lab Med. 2008;132:587–94.                       Infect Dis. 2000;6:601–8.
 3. Bugatti S, Manzo A, Caporal R, Montecucco C. Inflammatory             10. Dhingra KK, Gupta P, Saroha V, Setia N, Khurana N, Singh
    lesions in the bone marrow of rheumatoid arthritis patients: a mor-       T. Morphological findings in bone marrow biopsy and aspirate
    phological perspective. Arthritis Res Ther. 2012;14(6):229.               smears of visceral kala azar: a review. Indian J Pathol Microbiol.
 4. Travlos GS. Histopathology of the bone marrow. Toxicol Pathol.            2010;53:96–100.
    2006;34:566–98.                                                       11. Heegaard ED, Brown KE. Human parvovirus B19. Clin Microbiol
 5. Brackers de Hugo L, Ffrench M, Broussolle C, Sève                         Rev. 2002;15(3):485–505.
    P. Granulomatous lesions in bone marrow: clinicopathologic find-      12. Ganzel C, Constantin R. Parvovirus B19 diagnosed by bone mar-
    ings and significance in a study of 48 cases. Eur J Intern Med.           row biopsy. Blood. 2015;125:3351.
    2013;24(5):468–73.                                                    13. Alghamdi AA, Awan FS, Maniyar IH, Alghamdi NA. Unusual
 6. Diebold J, Molina T, Camilleri-Broet S, Le Tourneau A, Audouin            manifestation of extrapulmonary tuberculosis. Case Rep Med.
    J. Bone marrow manifestations of infections and systemic diseases         2013;2013:353798.
    observed in bone marrow. Trephine biopsy review. Histopathology.
    2000;37:199–211.
                                 Malignant Lymphomas
                                                                                                                                                     7
7.1       Introduction                                                3. Bone marrow biopsies may present the initial site of lymphoma
                                                                          diagnosis. In such instances, the patients are often noticed
Malignant lymphomas involving the bone marrow are sub-                    because of fever of unknown origin, abnormalities concerning
typed according to WHO classification [1–3]. In most                      the blood count or tumors in inaccessible localizations.
instances, a subdivision between Hodgkin lymphomas (HL)                4. After treatment of patients with bone marrow involve-
and non-Hodgkin lymphomas (NHL) as well as classifica-                    ment, BMBs are required for control purposes.
tion of malignant lymphomas according to WHO is possible
when immunohistochemistry is used. Ideally, additional flow               The infiltration pattern and the extent of neoplastic lym-
cytometric immunophenotypic studies, FISH, and molecular               phoid infiltrates can be exactly documented in BMBs. There
investigations are performed to estimate lineage, maturation           are two methods for estimation of the tumor cell burden:
stage, clonality, and genetic features of lymphoid cells.
However, the information gained from specialized tech-                 (a) the quantity of the lymphomatous infiltrate is stated as the
niques must be considered together with morphology and                     percentage of total nucleated cells with exclusion of fat cells
immunohistochemical results. PCR-studies for clonality                 (b) estimation of intertrabecular areas including adipocytes
concerning B- as well as T-lineage can show clonality in                   infiltrated by tumor cells
BMBs with benign lymphoid infiltrates and, the other way
around, PCR-studies may give negative results (germ line                  In Table 7.1, the typical morphologic features of BMBs
configuration) in malignant lymphomas [4]. Clonal T-cell               with infiltration of lymphomas are shown. Figure 7.1 reveals
populations can be detected in elderly patients and immuno-
compromised cases [5].
   The essential indications of bone marrow biopsies in
                                                                       Table 7.1 Bone marrow biopsies with suspected infiltration by malig-
malignant lymphomas are:                                               nant lymphomas: what has to be considered
                                                                        1. Infiltration: positive/   Staging and/or initial diagnosis
1. Bone marrow biopsies are performed as an integral part                  negative
   of routine staging in patients with a verified lymphoma              2. Subtyping of              Classification according to WHO
   diagnosis.                                                              lymphomatous
                                                                           infiltrate
2. In some instances, bone marrow biopsies are performed
                                                                        3. Infiltration pattern      Nodular, diffuse, interstitial, patchy,
   when suspicious, but not diagnostic lymphoid infiltrates                                          dense, intrasinusoidal
   are found in extramedullary localizations. The patholo-              4. Extent of infiltration    % area or % of nucleated cells
   gists know the diagnostic difficulties when tiny biopsy              5. Residual                  Quantity and quality
   material is sent from retroperitoneal tumors or other                   hematopoiesis
   localizations which are difficult to access and/or lym-              6. Bone structure         Increased remodeling
                                                                        7. Response to therapy Partial/complete remission, relapse,
   phoma is suspected clinically (e.g., unexplained spleno-
                                                                           in sequential biopsies concurrent disease
   megaly, lymphadenopathy, paraproteinemia, cytopenias).                                         (e.g., MDS, AL), toxic side-effects
   If the bone marrow is infiltrated by malignant lymphoma,             8. Transformation         High grade lymphoma, Hodgkin
   difficult surgical interventions are superfluous.                                              lymphoma
d e
Fig. 7.1 Schematic representation of infiltration patterns of malignant          (e) Diffuse infiltration pattern. (f) Patchy infiltration. (g) Dense infiltra-
lymphomas. (a) Nodular infiltration pattern. (b) Paratrabecular infiltra-        tion (packed marrow)
tion pattern. (c) Intrasinusoidal infiltration. (d) Interstitial infiltration.
7       Malignant Lymphomas                                                                                              133
f g
a schematic representation of the growth patterns of malig-     obscure the often subtle lymphoma infiltration. Therefore, it
nant lymphomas. However, a mixture of various growth pat-       is advisable to perform immunohistochemical stains using
terns in one biopsy of a patient can be seen.                   at least one B- and one T-cell marker (preferably CD20 and
   Differential diagnosis concerning reactive (benign) and      CD3).
neoplastic infiltrates must be carefully made especially in         In this chapter, characteristic and atypical examples of
cases with plasmacytosis (reactive or plasma cell myeloma),     BMBs with lymphoma infiltration are presented including
diffuse lymphocytosis or lymphoid nodules (reactive versus      clinical presentation, morphology, and immunohisto
malignant lymphoma) and granulomatous lesions (e.g., infec-     chemistry and, if necessary by PCR-techniques. Essential
tious disease or rheumatic diseases versus classical (c) HL).   antibodies currently used for correct classification are men-
   One has to be aware that lymphoma infiltration in the        tioned. We try to show the most important and rather com-
bone marrow can induce severe changes and alterations of        mon malignant lymphomas and the pitfalls; however, this
hematopoietic cells and the stroma. Such changes might          chapter cannot completely cover this issue.
134                                                                                                        7   Malignant Lymphomas
Case History 1 (CLL)                                                    lymphocytes: 52.8%. Spleen and liver showed normal size,
A 70-year-old male was admitted to the hospital because of              no enlarged lymph nodes were found. FACS-analysis was
recurrent cardiac problems. During clinical examination, a              suspicious of transformed lymphocytic lymphoma; there-
pathological blood count was noticed with leukocytosis and              fore, a bone marrow biopsy was performed (see Figs. 7.2
lymphocytosis: leukocytes: 12.2 G/L, neutrophils: 36.6%,                and 7.3).
Fig. 7.2 CLL (to case history 1). (a) Moderately hypercellular bone     are composed of small lymphocytes (H&E) (c–e) Infiltrating lympho-
marrow with multifocal nodular and patchy infiltration by lymphocytic   cytes show strong immunohistochemical reactivity for CD19 (c), CD20
lymphoma. The tumor burden is 30% (MGG). (b) Neoplastic infiltrates     (d), CD23 (e)
136                                        7   Malignant Lymphomas
CD19
b c
C D2 0
CD23
d e
CD5
                                                                                                                    MIB1
                                  CD3
b c
Fig. 7.3 CLL (to case history 1). (a) Infiltrating lymphocytes show       lymphoma characteristically reveals a low proliferation fraction (Ki67).
strong immunohistochemical reactivity for CD5. (b) Reactive CD3+          (d, e) Lymphoma cells in this case are CD38− (d) and ZAP-70- (e)
lymphocytes are found within the neoplastic infiltrate. (c) Lymphocytic
138                                        7   Malignant Lymphomas
                       CD38
                                  ZAP-70
d e
Case History 2 (CLL/SLL)                                                    lymphocytic lymphoma and associated with peripheral lym-
Tonsillar involvement by small lymphocytic lymphoma                         phocytosis, the diagnosis is CLL.
(SLL) was incidentally detected in a 31-year-old male                           A bone marrow biopsy was performed 10 days after the
patient. Bilateral tonsillectomy was performed because of                   diagnosis of tonsillar infiltration. Hematological work-up
chronic tonsillitis and tonsillar abscess. Both tonsils were                showed absolute B-lymphocytosis. Although the biopsy was
diffusely involved by SLL (see Fig. 7.4) with foci of resid-                markedly fragmented, nodular infiltration by lymphocytic
ual uninvolved tonsillar lymphoid tissue. Typical morpho-                   lymphoma, could be diagnosed (see Fig. 7.5a–f). However, a
logic features, such as small round lymphocytes and                         reliable statement concerning the tumor burden was not pos-
proliferation centers, were present and the neoplastic cells                sible due to fragmentation.
exhibited the characteristic CLL/SLL immunoprofile                              Diagnosis: fragmented bone marrow biopsy with nodular
(Fig. 7.4c–f): CD5+, CD20+, CD23+, negative for CD10                        infiltration by CLL.
and Cyclin D1.                                                                  Paraffin-embedded tissue from the bone marrow biopsy
   Diagnosis: Multifocal infiltration of both tonsils by                    was used for genetic analysis using NGS. Deletion of 17p13
SLL. Comment: if the bone marrow biopsy is infiltrated by                   was detected suggestive for an unfavorable diagnosis.
a b
c CD23 d CD23
Fig. 7.4 CLL (to case history 2 tonsil). (a, b) Infiltration of tonsil by   (MIB-1) is less than 5% within the neoplastic lymphocytic infiltrate. In
small lymphocytic lymphoma (H&E). (c, d) Tumor cells are positive           contrast, high proliferation fraction is seen in residual germinal centers
for CD23. (e) Strong positivity for CD5. (f) The proliferation fraction
140                                                                                                                7   Malignant Lymphomas
e CD5 f M IB 1
a b
c CD20 d CD5
Fig. 7.5 CLL (Case 2 bm). (a, b) Despite distinct fragmentation para-   to fragmentation). (c–e) Strong reactivity of infiltrating cells by CD20
trabecular and nodular infiltration can be seen by lymphocytic lym-     (c), CD5 (d), and CD23 (e). (f) Low proliferation fraction within neo-
phoma (MGG, H&E). Details about tumor burden are not possible (due      plastic cells in contrast to surrounding hematopoiesis (MIB-1)
7   Malignant Lymphomas                      141
e CD23 f M IB 1
a b
c d
Fig. 7.6 CLL—Richter’s syndrome (to case history 3; transformation         The infiltrates of CLL and the tumor cells of transformed lymphoma
to Richter’s syndrome). (a–d) Hypercellular bone marrow with nodular       reveal an identical phenotype with positivity for CD19 (e), CD20 (f),
infiltrates of typical CLL and nodular to patchy infiltration by diffuse   CD23 (g), and CD5 (h)
large B-cell lymphoma according to Richter’s syndrome (H&E). (e–h)
7    Malignant Lymphomas                                                                                                  143
CD19 e CD20 f
CD23 g CD5 h
Case History (PLL)                                                          Diagnosis: Highly hypercellular bone marrow with diffuse
A 76-year-old male was admitted to the hospital because                 infiltration by B-cell prolymphocytic leukemia. The percent-
of splenomegaly. Laboratory investigation revealed                      age of tumor infiltration compared to hematopoiesis is 80%.
marked leukocytosis (85.809/L) and lymphocytosis                            At the same time as the bone marrow biopsy was per-
(95%). Furthermore, cervical lymphadenopathy (1 cm in                   formed, a cervical lymph node was removed for histological
diameter) was observed. Blood biochemistry was normal                   investigation (Fig. 7.8)
except for elevated lactate dehydrogenase level with                        The lymph node showed a diffuse infiltration by lympho-
546 U/L (normal: 120–140 U/L). A bone marrow biopsy                     cytes identical to those in the bone marrow, (Fig. 7.8a–c).
(see Fig. 7.7) and lymph node extirpation (see Fig. 7.8)                Similar to the bone marrow, the tumor cells were strongly
were performed.                                                         CD20+ (Fig. 7.8d). Many reactive CD3+ T-cells were intermin-
                                                                        gled (Fig. 7.8e). Using CD5 antibody more cells were stained
    Bone marrow biopsy revealed highly hypercellular                    than with CD3, implicating that both T-cells and tumor cells
bone marrow with diffuse infiltration by medium-sized                   expressed CD5 (Fig. 7.8f). In the lymph node, most of the infil-
lymphocytes with round nuclei and moderately dense                     trating cells were CD23+ (Fig. 7.8g). The proliferation fraction
 chromatin as well as centrally located nucleoli (Fig. 7.7a–            using MIB1 in the lymph node was about 20% (Fig. 7.8h).
 c). Immunohistochemical investigations showed strong                       Diagnosis: Lymph node infiltration by B-cell prolympho-
 expression of CD19, CD20 (Fig. 7.7d), CD22, and                        cytic leukemia.
 CD79a. About 30% of these tumor cells were positive for                    Genetic analyses on paraffin-embedded lymph node tis-
 CD23 (Fig. 7.7e), but there was no reactivity for CD5.                 sue were performed using next-generation sequencing
 The proliferation fraction (Fig. 7.7f) using MIB1 ranged               (NGS) detecting trisomy 12, a rather uncommon cytogenetic
 from 10 to 15%.                                                        abnormality in B-PLL.
a b
Fig. 7.7 PLL (bone marrow). (a, b) (B-PLL). Highly hypercellular        for CD20. (e) A small proportion of tumor cells is CD23+. (f)
bone marrow with diffuse infiltration by B-PLL (MGG). (c) Most of the   Proliferation fraction using MIB1 is 10–15%
tumor cells show nucleoli (H&E). (d) Strong reactivity of tumor cells
146                                                                                                             7   Malignant Lymphomas
c CD20 d
CD23 e MIB1 f
a b
Fig. 7.8 PLL (lymph node). (a–c) Dense lymph node infiltration by     tumor cells show CD5-positivity. (g) In the lymph node, more tumor
B-PLL (H&E). (d) Strong reactivity of tumor cells using antibody to   cells are CD23+ than in the bone marrow. (h) Proliferation fraction is
CD20. (e) Many reactive T-lymphocytes are interspersed (CD3). (f)     about 20% (MIB1)
Compared to CD3, in addition to reactive T-lymphocytes, also some
7    Malignant Lymphomas                                                                                             147
c CD20 d
CD3 e CD5 f
CD23 g MIB1 h
7.4.2    Morphology
                                                                    7.4.4   Cytogenetic Abnormalities
LPL shows a mixture of small lymphocytes, lymphocytes with                   and Molecular Characteristics
plasmacytoid differentiation and plasma cells. Russell bodies
corresponding to cytoplasmic or extracellular i mmunoglobulin      NGS studies have detected somatic mutations of MYD88, a
deposits, as well as intranuclear inclusions, referred to as        key component of the Toll-like receptor signaling machin-
Dutcher bodies can be present. Some blasts can be intermin-         ery, in most cases of LPL/WM. Deregulated MYD88 signal-
gled. The number of these cells is variable. Occasionally,          ing promoted by mutations support tumor cell survival in
plasma cells are the predominant cells, especially in treated       LPL/WM, demonstrating that they are gain-of-function
LPLs. Distinction from plasma cell myeloma can be difficult         driver events in this lymphoma [29]. Otherwise, no specific
morphologically and may require immunohistochemical and             chromosomal or oncogenic abnormalities are found in
molecular investigations. Usually, many mast cells can be seen      LPL. Frequently, deletion 6q is reported in LPL; however,
between the lymphoma cells or surrounding infiltrates. LPL         this finding is not specific. Translocations involving the
involves the bone marrow in a nodular, diffuse, and/or intersti-    immunoglobulin heavy chain locus occasionally are
tial pattern and paratrabecular infiltrates are frequently found.   reported [30].
7   Malignant Lymphomas                                                                                                                  149
Case History 1 (LPL)                                                       lymph node involvement by LPL, a bone marrow biopsy was
A 65-year-old woman was admitted to the hospital for strip-                performed showing lymphoma infiltration (Fig. 7.9a–h).
ping operation of right vena saphena magna. During this                    DNA extracted from paraffin-embedded material of the bone
operation, a small (diameter 9 mm) inguinal lymph node was                 marrow biopsy revealed a MYD88 L265P mutation.
removed. Clinical records of the patient, who was treated for
osteoporosis since 13 years, revealed monoclonal gammopa-                      Diagnosis: Highly hypercellular bone marrow with dense
thy of IgM lambda type since 9 years. Complete blood count                 infiltration by LPL/Waldenström macroglobulinemia with
and differential blood count were normal; however, serum                   expression of IgM kappa. Tumor burden, in relation to hema-
protein kappa was highly elevated. After the diagnosis of                  topoiesis was more than 90%.
a b
c d
Fig. 7.9 Case history 1 (LPL). (a–c) Highly hypercellular bone mar-        Russell bodies are lambda-negative. (g) The lymphocytes are CD20-
row with dense infiltrate of lymphocytes, plasma cells, and Russell bod-   positive. (h) Strong reactivity of lymphoplasmacytic infiltrate and
ies (H&E). (d) Delicate fibrosis is detected by Gomori’s stain. (e)        Russell bodies with an antibody to IgM
Expression of immunoglobulin kappa. (f) The infiltrating cells and
150                                7   Malignant Lymphomas
e f
kappa lambda
g h
CD20 IgM
Case History 2 (LPL/Waldenström Figs. 7.10, 7.11,                        tion of Codon 265 (exon 5) of the MYD88 gene was detected
and 7.12)                                                                in the DNA extracted from the bone marrow biopsy.
A 74-year-old woman was admitted to the hospital to undergo
a thorough diagnostic work up because of anemia and slight                  Diagnosis: Highly hypercellular bone marrow with multifo-
splenomegaly as well as monoclonal gammopathy of IgM                     cal nodular and patchy infiltration by LPL with expression of
lambda type. Furthermore, slightly enlarged axillary lymph               IgM lambda, consistent with Waldenström macroglobulinemia.
nodes (Fig. 7.10) were found. A bone marrow biopsy                       The tumor burden was estimated as 40% of nucleated cells.
revealed multifocal infiltration by LPL (Fig. 7.11). A muta-
a b
c d
CD20
Fig. 7.10 Lymph node (case history 2). (a–c) H&E stains of the ingui-    nodules, suggestive of residual follicles, are detected. (e) Plasma cells
nal lymph node with blurred architecture and focal sinus histiocytosis   are negative for light chain kappa. (f, g) Lambda-positive plasma cell
(a, b). High magnification shows increased number of plasma cells and    population. (h) The plasma cells express IgM heavy chain
occasional Dutcher bodies (c). (d) With an antibody to CD20 B-cell
152                                                                                                               7   Malignant Lymphomas
e f
kappa lambda
g h
lambda IgM
a b
Fig. 7.11 Bone marrow (case history 2). (a–d) Highly hypercellular     Infiltrate stained with H&E showing some Dutcher bodies. (g) Dutcher
bone marrow with multifocal nodular to patchy neoplastic lymphoplas-   body (center) highlighted by PAS-stain. (h) Within the neoplastic infil-
macytic infiltrates with many interspersed mast cells (MGG). (e, f)    trate a delicate fibrosis is found (Gomori’s stain)
7       Malignant Lymphomas       153
c d
e f
g h
a b
kappa kappa
c d
lambda lambda
e f
CD20 CD23
Fig. 7.12 (a, b) Lymphocytes and plasma cells are kappa-negative. (c,           an antibody to CD23. (g) The many interspersed mast cells are stained
d) Strong lambda-positivity of infiltrating cells. (e) Majority of infiltrat-   with an antibody to CD117c. (h) Low proliferation fraction of the infil-
ing cells show CD20-positivity. (f) Many tumor cells are reactive with          trating cells (MIB-1)
7       Malignant Lymphomas                                                                                                155
g h
CD117c MIB1
7.5      Mantle Cell Lymphoma (MCL)                              morphology closely resembles other small B-cell lympho
                                                                  proliferative diseases, such as chronic lymphocytic leukemia,
7.5.1    Epidemiology                                            prolymphocytic leukemia, follicular lymphoma, and mar-
                                                                  ginal zone lymphoma. Correct diagnosis is only possible with
The reported incidence of MCL is 3–10 % of all NHLs with          immunophenotyping. In the blastoid variant of MCL, the
a marked male predominance (2:1 or even greater) [1–3].           lymphoma cells mimic large cell lymphoma or lymphoblastic
Rarely, patients under the age of 50 are affected; the median     leukemia revealing tumor cells with dispersed chromatin,
age at first diagnosis is about 65 years [31]. Although there     small nucleoli, and scant cytoplasm. In blastoid variants of
are reports detecting skewed immunoglobulin variable              MCL, the infiltration pattern is either diffuse or interstitial.
gene segment usage stimulating lymphomagenesis, a con-
clusive link to autoimmune disorders or pathogens could
not be identified until now [31]. MCL may be an aggressive        7.5.3    Immunohistochemistry
disorder with a fast-growing tumor mass, but it can also be
rather indolent in some patients, especially in those, show-      In contrast to B-CLL, cells of MCL are strongly CD20+ and
ing exclusively blood and bone marrow involvement. The            show a rather strong surface immunoglobulin light chain
disease is called “mantle cell lymphoma” because the              restriction. Usually, cells of MCL are CD23-negative; how-
tumor cells originate from the mantle zone of the lymph           ever, weak expression of CD23 is possible. Characteristically,
node. MCL is usually diagnosed as a late-stage disease            the tumor cells are CD5-positive and CD10 as well as BCL6-
with multiple organ involvement. MCL confined to the gas-         negative. Almost all cases are Cyclin D1+ and all cases are
tro-intestinal tract is referred to as lymphomatoid polyposis     BCL2+. Classical cases of MCL are SOX11 (a transcription
(Fig. 7.13).                                                      factor) positive (nuclear staining), blastoid variants of MCL
                                                                  are frequently SOX11-negative [34].
7.5.2    Morphology
                                                                  7.5.4    Cytogenetic Abnormalities
The pattern of bone marrow involvement is rather variable                   and Molecular Characteristics
from nodular, interstitial, and paratrabecular to diffuse [32].
Occasionally, prominent intrasinusoidal infiltration has been     The translocation t(11;14) can be detected in more than 90%
observed in MCL [33].                                             of MCLs.
   Mantle cell lymphoma has a high frequency for marrow              Alterations of the TP53 locus at chromosome 17p13 are
involvement (up to 95%) with small lymphoid cells, some           frequently found in MCL. In about 20% of patients, muta-
with round and others with indented or angulated nuclei.          tions of TP53 have been reported and these patients are
Reticulin fibers are mostly increased in infiltrated areas. The   known to be associated with a rather poor outcome [35].
7   Malignant Lymphomas                                                                                                                        157
Case History 1; MCL in colonic biopsies (Fig. 7.13) and                     Colonic biopsies showed infiltration by mantle cell lym-
bone marrow biopsy (Fig. 7.14)                                              phoma (Fig. 7.13a–h). A bone marrow biopsy was per-
A 75-year-old male was admitted to the hospital because of                  formed. This biopsy was infiltrated by mantle cell lymphoma
obscure abdominal pain. The diagnostic work-up revealed                     in a paratrabecular and interstitial pattern (see Fig. 7.14). The
slight splenomegaly, slight anemia, and thrombocytopenia.                   tumor burden in relation to hematopoiesis was 30%.
a b
c Cyclin D1 d
CD21 e CD21 f
Fig. 7.13 MCL Colon. (a–b) Colonic biopsy with nodular and patchy           (e, f) Within the infiltrate, dense irregular meshwork of FDCs is pres-
infiltration by MCL. (c) High magnification of the infiltrate shows         ent. (g) Reactive CD3-positive lymphocytes are intermingled. (h)
the typical cleaved tumor cells. (d) Strong nuclear Cyclin D1-positivity.   Lymphoma cells are CD5-positive
158                                                                                                                  7   Malignant Lymphomas
CD3 g CD5 h
a b
c d
Fig. 7.14 MCL bone marrow. (a–d) Accentuated paratrabecular lym-            Coexpression of CD5 in the lymphoma cells. (h) In most tumor cells,
phoma infiltration (H&E). (e, f) CD20-positive tumor cells are infiltrat-   nuclear staining for Cyclin D1 is seen (inset shows another area with
ing the bone marrow in a paratrabecular and interstitial pattern. (g)       Cyclin D1-positive cells)
7    Malignant Lymphomas                                                                                               159
CD20 e C D20 f
CD5 g Cyclin D1 h
a b
c d
CD20 CD23
e f
Cyclin D1 Cyclin D1
Fig. 7.15 In situ MCL. (a, b) Closely attached to the bony trabeculae,       reactive BCL6 (left side) positive and CD10 (right side) positive germi-
a lymphoid follicle is found (H&E). (c) Staining with an antibody to         nal center cells. (h) High proliferation fraction in the reactive germinal
CD20 shows a rather “normal” looking follicle. (d) A concentric net-         center and rather low proliferation fraction in the neoplastic mantle
work of CD23-positive follicular dendritic cells is seen in this follicle.   zone (MIB1)
(e, f) Cyclin D1-positive mantle zone cells. (g) The follicle contains
7   Malignant Lymphomas                       161
g h
7.6      Hairy Cell Leukemia (HCL)                                 mast cells, plasma cells, small lymphocytes, a variable number
                                                                    of hematopoietic precursors, and typically extravasated eryth-
7.6.1    Epidemiology                                              rocytes [36]. Hematopoiesis, especially erythrocytopoiesis, in
                                                                    most instances shows dysplastic features. In most cases, the
HCL is a rare mature B-cell neoplasm accounting for 2% of           bone marrow biopsy is hypercellular, however, hypocellular
lymphoid leukemias. The median age is 52 years with a               biopsies, resembling aplastic anemia, may occur [37].
male: female ratio of 5:1.
                                                                    7.6.3    Immunohistochemistry
7.6.2    Morphology
                                                                    The characteristic immunophenotype of HCL shows expres-
Hairy cells are medium-sized lymphocytes, about twice the           sion of CD19, CD20, CD22, CD79a, and CD11c and addi-
size of normal small lymphocytes. The nuclei are round, oval,       tional expression of CD103, CD25, CD123, Annexin A1,
bean-shaped, or convoluted with a rather homogenous chro-           BRAF, TRAP (tartrate-resistant acid phosphatase), DBA44,
matin. Rarely, the nuclei are bilobulated. If nucleoli are pres-    FMC-7, and variably Cyclin D1 [38].
ent, these are inconspicuous. The cytoplasm is broad and
irregularly shaped with lateral interdigitating extensions
(“hairy” projections are only seen in smears). Due to these         7.6.4    Cytogenetic Abnormalities
projections, the infiltration consists of widely separated cells;             and Molecular Characteristics
the cells almost never show overlapping in normal thickness
sections. The bone marrow infiltration is highly variable, rang-    There is no specific cytogenetic abnormality for
ing from a minimal interstitial infiltrate to obvious diffuse to    HCL. Numerical abnormalities of chromosomes 5 and 7
even solid infiltration. The hairy cells are usually dispersed      have been described [39]. The majority of cases of HCL
within a reticular fiber network. The clinician thus often gets a   show VH genes with somatic hypermutation [39]. Usually,
dry tap and the bone marrow biopsy is the only reliable tech-       BRAF-V600E mutation as the disease-defining genetic event
nique for confirming the diagnosis. The infiltrate also contains    in HCL can be detected [40].
7   Malignant Lymphomas                                                                                                                    163
Case History 1; HCL                                                         The bone marrow was slightly hypercellular for age. This
An 81-year-old male was admitted to the hospital because of              was due to a diffuse infiltration of atypical lymphocytes
progredient pancytopenia, monocytopenia (Leuko 2.70 G/l,                 which exhibited the immunophenotype of HCL. The tumor
Ery 3.8 T/L, Thrombo 55 G/l, Mono 1%), and splenomegaly                  burden in relation to hematopoiesis was 50%.
(18 cm). No enlarged lymph nodes were found. In the periph-                 The diagnosis of diffuse bone marrow infiltration by HCL
eral blood, hairy cells were seen, FACS-analysis was suspi-             was straightforward (see Figs. 7.16 and 7.17) with a tumor
cious of clonal B-cell proliferation, consistent with HCL. A             burden of 50%.
bone marrow biopsy was performed (Figs. 7.16 and 7.17).
a b
c d
e f
CD19 CD19
Fig. 7.16 Case 1 (HCL) Slightly hypercellular bone marrow for age        investigations reveal diffuse bone marrow infiltration by typical HCL.
(a–c). In higher magnification (d), the atypical lymphocytic cells are   (e, f) CD19-positive cells; (g, h) strong CD20-positivity
surrounded by extravasated erythrocytes. (e–h) Immunohistochemical
164                                                                                                                    7   Malignant Lymphomas
g h
CD20 CD20
a b
CD11c CD11c
c d
CD25 CD103
Fig. 7.17 Case 1 (HCL) (a–d) Immunohistochemical investigations           detectable. (f) Megakaryocytes, stained with an antibody to CD61, are
reveal diffuse bone marrow infiltration by typical HCL. (a, b) Specific   seen diffusely dispersed. (g, h) Diffuse reticulin fiber fibrosis (Gomori’s
staining for CD11c; (c, d) Tumor cell are CD25 and CD103-positive.        stain) is typically seen between the HCL cells
(e) Between tumor cells erythropoietic islands (stained with CD71) are
7       Malignant Lymphomas                165
e f
CD71 CD61
g h
GOM GOM
a b
c d
e f
MPO CD61
Fig. 7.18 Case 2 (HCL). (a, b) Hypercellular bone marrow with dif-           MPO (e) stains residual granulopoietic cells, CD61 (f) residual mega-
fuse infiltration of atypical lymphocytes. In higher magnification (c, d),   karyocytes and CD71 (g) residual erythropoiesis. (h) The proliferation
the lymphocytes show an ample cytoplasm and slightly irregular nuclei        fraction within the tumor cell population is very low, only erythropoi-
with inconspicuous nucleoli. (e–g) Highly reduced hematopoiesis.             etic cells show a high proliferation fraction using MIB1
7       Malignant Lymphomas                                                                                                                   167
g h
CD71 MIB1
a b
CD20 CD11c
c d
CD25 CD103
Fig. 7.19 Case 2. (a–d) Tumor cells of HCL are reactive with antibod-   specifically stained with an antibody to light chain lambda (f). (g) Cells
ies to CD20 (a), CD11c (b), CD25 (c), and CD103 (d). (e, f) Using an    of HCL are negative for CD5. (h) Single hairy cells show reactivity
antibody to light chain kappa, cells of HCL are negative, only inter-   with an antibody to CD23
mingled plasma cells are reactive (e). In this case, cells of HCL are
168                                                                                                    7   Malignant Lymphomas
e f
kappa lambda
g h
CD5 CD23
7.7       Follicular Lymphoma (FL)                                         usually three times the size of lymphocytes and reveal one or
                                                                            more, often peripherally located nucleoli. It is not uncommon
7.7.1     Epidemiology                                                     that the bone marrow biopsy is infiltrated by “low grade“(grade
                                                                            1 and 2) FL, while the lymph node of the affected patient shows
FL approximately accounts for 20% of all lymphomas and is                   “high grade” FL (grade 3A/B or DLBCL). Grading of FL in
the most frequent type of lymphoma in Western countries.                    bone marrow biopsies is challenging since often small numbers
Mainly adults with a median age in the sixth decade are                     of tumor cells are present. In rare instances, FL reveals a signet
affected, the male: female ratio is 1:1.7. Rarely, FL occurs                ring cell morphology (Fig. 7.20c, d), a feature which can also
under the age of 20 years and if FL occurs in pediatric patients            be evident in extranodal marginal zone B-cell lymphomas,
(pediatric FL), the affected children are male. There is a high             lymphocytic lymphomas, diffuse large B-cell lymphomas, or
incidence of bone marrow infiltration in “low grade” FL with                plasma cell myelomas [43]. In most cases, the bone marrow
50–60% and in “high grade” FL with about 30% [42].                          shows an accentuated paratrabecular either nodular, patchy,
                                                                            band-like or mixed infiltration pattern (Fig. 7.20e, f). In cases
                                                                            of band- like paratrabecular infiltrates, the lymphoma cells
7.7.2     Morphology                                                       often are barely visible since only delicate rims of tumor cells
                                                                            are attached to the trabecular bone (Fig. 7.20g, h). Other cases
As in lymph nodes, FL in the bone marrow is composed of                     show extensive broad rims of paratrabecular infiltrates. In addi-
centrocytes and centroblasts (Fig. 7.20a, b). Centrocytes are               tion, FL can show an interstitial, diffuse, or nodular growth
small- to medium-sized cells with cleaved or angulated nuclei,              pattern. In rare instances, a true follicular growth pattern with a
inconspicuous nucleoli, and scant cytoplasm. Centroblasts are               meshwork of follicular dendritic cells can be recognized [44].
a b
c d
Fig. 7.20 FL. (a) Follicular infiltration by FL. (b) Higher magnifica-      FL (H&E). (f) Band-like paratrabecular infiltration by FL (H&E).
tion shows centroblasts and centrocytes in germinal center like struc-      (g) In H&E stain, the delicate rim of lymphoma cells easily can be
ture (H&E). (c) Patchy paratrabecular infiltration by FL with signet ring   missed. (h) The delicate rim of the lymphoma infiltrate is highlighted
cell appearance (H&E). (d) Higher magnification of signet ring cell like    by immunohistochemical stains using an antibody to CD20
FL (H&E). (e) Marked nodular to patchy paratrabecular infiltration by
170                                                                                             7   Malignant Lymphomas
e f
g h
CD20
Case History 1 (FL)                                                        (grade 2). For staging reasons, a bone marrow biopsy was per-
A 46-year-old woman presented with enlarged cervical and                   formed. This biopsy showed slight to moderate hypercellular
axillary lymph nodes. The liver and spleen were of normal size,            bone marrow with multifocal nodal and patchy paratrabecular
the blood count was also normal. An axillary lymph node was                and follicular infiltration by “low grade” FL (see Fig. 7.21).
surgically removed and revealed infiltration by “low grade” FL             The tumor burden, in relation to hematopoiesis was 25%.
a b
CD20
c d
CD23 CD23
e f
CD21 CD10
Fig. 7.21 Case history 1 (FL). (a) Marked paratrabecular nodular to        infiltrates show networks of follicular dendritic cells (CD21). (f) In the
patchy infiltrates by FL (H&E). (b) The infiltrates are clearly visible    follicular infiltrates, the tumor cells are CD10+, whereas in paratrabec-
using an antibody to CD20. (c, d) The paratrabecular infiltrates and the   ular infiltrates only single cells are CD10+. (g) The lymphoma cells are
follicular non-paratrabecular infiltrates express CD23. (e) Some of the    strongly BCL2+. (h) Many, but not all tumor cells express BCL6
172                                                                                                      7   Malignant Lymphomas
g h
BCL2 BCL6
a b
c d
MGG MGG
Fig. 7.22 SMZL. (a, b) Slightly hypercellular bone marrow with inter-       intrasinusoidal infiltration is easily detected (f). (g, h) Staining of inter-
stitial and intrasinusoidal infiltration by SMZL (H&E). (c, d) In MGG       stitial and intrasinusoidal lymphocytic infiltrate with B-cell markers
stained sections, interstitial and intrasinusoidal small lymphocytes are    (g: CD19, h: CD20)
visible. (e) No reticulin fibrosis is seen (Gomori’s stain), however, the
7       Malignant Lymphomas                                                                                                                       175
e f
GOM GOM
g h
CD19 CD20
a b
CD20 CD20
Fig. 7.23 SMZL. (a, b) Higher magnification of intrasinusoidal               cytic cells are CD103-positive. (g, h) Intermingled reactive CD3-positive
CD20-positive lymphocytic cells. (c, d) The infiltrating cells are kappa-   T-cells, focally arranged in a nodular pattern
positive (c) and negative for lambda (d). (e, f) The neoplastic lympho-
176                                 7   Malignant Lymphomas
c d
kappa lambda
e f
CD103 CD103
g h
CD3 CD3
a b
CD5 CD10
         c
                                                                            d
MIB1 MIB1
e f
Fig. 7.24 SMZL. (a, b) The infiltrating cells are not reactive with anti-   ality (light chain Ig kappa B) is verified (285 nt). (f) PCR: primers for
bodies to CD5 (a) and CD10 (b). (c, d) Using a proliferation marker         heavy chain gene: clonal peak is seen in DH (252 nt)
(MIB1), mainly erythroid cells are stained. (e) With PCR method, clon-
The lymphoma cells show small- to medium-sized slightly          7.9.4   Cytogenetic Abnormalities
irregular nuclei with inconspicuous nucleoli similar to cen-              and Molecular Characteristics
trocytes. Occasionally, the lymphoma cells reveal a mono-
cytic appearance with a pale, rather abundant cytoplasm. A       There are varying translocations or trisomies depending on
plasmacytic differentiation can be obvious. A few blasts are     the site of primary disease. Thus, t(11;18)(q21;q21) is most
intermingled. In most cases, the bone marrow is infiltrated in   often detected in gastric and pulmonary lymphomas, t(14;18)
a nodular pattern; however, an interstitial or combined nodu-    (q32;q21) is mainly detected in skin, ocular, and thyroid
lar/interstitial infiltration pattern can be seen. Rarely, an    MALT lymphomas.
intrasinusoidal lymphoma spread can be found [55].
7       Malignant Lymphomas                                                                                                                       179
Case History 1 (MALT); Fig. 7.25                                            (see Fig. 7.25c). IgH rearrangement studies showed identical
A 76-year-old male was admitted to the hospital because of                  clonal peaks in the soft palate and the bone marrow.
a slow growing swelling at the soft palate. A biopsy                           Diagnosis: Normocellular bone marrow with multifocal
(Fig. 7.25a, b) revealed infiltration by an extranodal MALT                 nodular infiltration by a low grade B-cell lymphoma, consis-
lymphoma. Staging procedures, including a bone marrow                       tent with MALT lymphoma. The percentage of the infiltra-
biopsy, showed a nodular paratrabecular infiltration by a low               tion is 5%.
grade B-cell lymphoma, consistent with “MALT lymphoma”
a b
c d
Fig. 7.25 Case history 1. MALT. (a, b) Infiltration of the soft palate by   palate. (e, f) Follicular dendritic reticulum cells are seen in the lym-
a low grade lymphoma with intermingled reactive and colonized germi-        phoma of the soft palate (e) and also in the neoplastic infiltrates of the
nal centers (e reveals CD21-positive FDCs in these germinal centers         bone marrow (f). (g, h) Strong reactivity of the infiltrating cells with an
according to a “MALT” lymphoma). (c, d) Morphology of the nodular           antibody to CD20 and light chain restriction (tumor cells are kappa-
infiltrates in the bone marrow is similar to the tumor cells in the soft    positive [left] and lambda-negative [right])
180                                        7   Malignant Lymphomas
e f
CD21 CD23
g h
Case History 2 (MALT) Fig. 7.26                                              lesions in both lungs. Histologic examination of the lesions
A 35-year-old woman was diagnosed with Helicobacter                          in stomach, liver, and lung confirmed the diagnosis of gen-
pylori-positive MALT lymphoma. HP eradication therapy                        eralized MALT lymphoma. Furthermore, infiltration by
was performed without any staging procedures. Since con-                     this lymphoma was seen in the bone marrow biopsy.
trol endoscopy after 6 months still revealed lymphoma, the                       Diagnosis: Normocellular bone marrow with nodular
patient was referred to the hospital. Computed tomography                    infiltration by “low grade” B-cell lymphoma, consistent with
scans revealed a diffuse gastric infiltration of the antrum                  an infiltration of the already verified extranodal MALT lym-
and corpus, enlargement of epigastric lymph nodes, as                        phoma. The percentage of the infiltration is 10% of nucleated
well as an 8 × 7 cm hypodense liver lesion and multiple                      cells.
a b
c d
Fig. 7.26 Case history 2. MALT (a, b) show the infiltration in the liver     infiltrating cells in the bone marrow biopsy. (f) The proliferation frac-
by small centrocyte-like cells with occasional intermingled blasts. (c, d)   tion (MIB 1) is less than 5% in the lymphoma population. (g, h)
Nodular infiltration of the bone marrow by an identical lymphoma pop-        Lymphoma cells reveal positivity for kappa light chain, whereas the
ulation as shown in the liver (H&E). (e) Strong CD20-positivity of the       cells are negative using an antibody to lambda light chain
182                                                                                           7   Malignant Lymphomas
e f
CD20 MIB1
g h
kappa lambda
Case History 1 (DLBCL)                                                      Diagnosis: Highly hypercellular bone marrow with dense
A 58-year-old previously healthy woman presented with                   infiltration by a “high grade” B-cell lymphoma. The mor-
dyspnea, retrosternal pain, and increased tendency to                   phology and immunohistochemical results are consistent
bleed. Examination revealed splenomegaly (diameter                      with infiltration by DLBCL; immunohistochemical algo-
15 cm) and pancytopenia with marked anemia (Hb 6.9 g/                   rithm: activated B-cell like, immunohistochemical double hit
dL, Hkt17.8%). A bone marrow biopsy was performed.                      score according to Green: 2. The percentage of infiltration in
This biopsy revealed a dense infiltration by DLBCL of                   relation to hematopoiesis is >90%.
ABC subtype (Fig. 7.27).
a b
c d
CD20 BCL2
Fig. 7.27 (a, b) The highly hypercellular bone marrow shows dense       the blasts are c-MYC-positive; strong positivity is seen with an anti-
infiltration by lymphoid blasts (H&E). (c, d) The blasts are strongly   body to BCL6. (g, h) The blasts show intranuclear FOXP1-positivity
reactive with antibodies to CD20 and BCL2. (e, f) More than 30 % of     and a high proliferation fraction (Ki67)
7       Malignant Lymphomas                 185
e f
cMYC BCL6
g h
FOXP1 Ki 67
a b
CD20
c d
CD3 GCET1
Fig. 7.28 (a) Highly hypercellular bone marrow with multiple nodular          the blasts with an antibody to GCET1. (e) Using an antibody to CD10
and patchy, occasionally paratrabecular located infiltrates by blasts         only dendritic cells are stained, the tumor cells are negative. (f) Only
(inset shows the blasts with one or more nucleoli). (b) The infiltrates are   some blasts are weakly BCL6-positive. (g) Strong FOXP1-positivity of
highlighted by staining with an antibody to CD20. (c) Reactive CD3-           the blasts. (h) The proliferation fraction (MIB1) is higher than 80%
positive T-lymphocytes are intermingled. (d) There is no reactivity of
7   Malignant Lymphomas                                                                                              187
e f
CD10 BCL6
g h
FOXP1 MIB1
7.11       Intravascular Large B-Cell Lymphoma                               Case History (intravascular large B-cell lymphoma, see
                                                                              Fig. 7.29)
This is an aggressive lymphoma in which the tumor cells                       An 82-year-old woman was admitted to the hospital because of
grow within vascular lumina of various organs including                       marked anemia. The patient had no enlarged lymph nodes and a
the bone marrow (see Fig. 7.29), which can be the initial                     normal size of spleen and liver. Since the anemia could not be
site of diagnosis [62, 63]. Because this type of lymphoma is                  explained, a bone marrow biopsy was performed. In a moderate
extremely rare, no detailed descriptions concerning epide-                    to highly hypercellular bone marrow, the sinusoids and some
miology, morphology, and cytogenetic abnormalities are                        small vessels were filled with huge B-blasts corresponding to an
available.                                                                    infiltration by intravascular large B-cell lymphoma (Fig. 7.29).
    In bone marrow sections, aggregates of blasts are seen in                     Diagnosis: Highly hypercellular bone marrow with infil-
vessels, especially sinusoids. These blasts can be highlighted                tration by intravascular large B-cell lymphoma.
by immunohistochemical stains with B-cell markers. More
than 30% of such lymphomas show expression of CD5,
about 10% reveal expression of CD10. In cases with CD10- 7.11.1 Caution
negativity, the blasts, like in our case shown below, are
MUM1-positive.                                                 Minimal extravascular infiltration may be seen in some cases
                                                               of intravascular large B-cell lymphoma.
a b
c d
CD20 CD20
Fig. 7.29 (a) The bone marrow is highly hypercellular with only a few         antibody to CD34 labels endothelial cells thus highlighting the intravas-
fat cells. (b) Sinusoids and small vessels are filled with huge blasts. (c,   cular blast infiltration. (f) Most blasts express CD5. (g, h) The blasts are
d) The intravascular blasts are positive for CD20. (e) Staining with an       CD10-negative (g) and MUM1-positive (h)
7   Malignant Lymphomas                 189
e f
CD34 CD5
g h
CD10 MUM1
7.12     Burkitt Lymphoma (BL)                                   mingled macrophages give rise to the typical “starry sky”
                                                                  appearance.
7.12.1 Epidemiology
7.12.2 Morphology
                                                                  7.12.4 Cytogenetic Abnormalities
The bone marrow, which is infiltrated in more than half of the            and Molecular Characteristics
patients, shows an interstitial or diffuse infiltration pattern
(see Fig. 7.30e, f). The tumor cells are of medium size with      The majority of Burkitt lymphomas reveal a MYC transloca-
multiple small nucleoli. The characteristic intracytoplasmic      tion at band 8q24 to the IG heavy chain region 14q32. A few
vacuoles are better visualized in smears or touch prepara-        cases have MYC translocation at the lambda 22q11 or kappa,
tions. Many mitoses and apoptosis are seen. Numerous inter-       2p12 light chain loci [65].
7   Malignant Lymphomas                                                                                                                    191
Case History (Burkitt Lymphoma)                                           lymphoma (Fig. 7.30a–d). The monomorphic blasts, infil-
A 29-year-old patient initially presented with acute abdo-                trating the small bowel, revealed high nuclear to cytoplasmic
men, leucocytosis 15.8 × 103/μL, thrombocytopenia 72 × 103/               ratio and two to three nucleoli. The proliferation fraction was
μL, and mild anemia. Emergency operation revealed perfora-                100%. Bone marrow biopsy showed diffuse infiltration by
tion of the small bowel due to a large solid tumor mass. In               c-MYC-positive Burkitt lymphoma (Fig. 7.30e, f).
H&E sections, the tumor masses showed blasts of Burkitt
a b
c d
MIB1
Fig. 7.30 (a) Dense infiltration of the small bowel by Burkitt lym-       Routinely stained H&E section shows diffuse blastic infiltration of the
phoma. (b) Characteristic “starry sky” pattern of this lymphoma. (c) At   bone marrow. (f) Immunohistochemical staining reveals c-MYC-
higher magnification, monomorphic blasts with several nucleoli are        positivity in most blasts. (g) MYC translocation at band 8q24 to the IG
seen. (d) Proliferation fraction using MIB1-antibody is 100%. (e)         heavy chain region could be demonstrated by FISH analysis
192                                                                                        7   Malignant Lymphomas
e f
c-myc
The expression “T-CLL” is used as a synonym for small cell       7.13.4 Cytogenetic Abnormalities
variants of T-PLL. The leukemia is characterized by the                  and Molecular Characteristics
proliferation of small- to medium-sized lymphoid cells with
 non-granular basophilic cytoplasm and small visible nucle-      Clonal rearrangement for T-cell receptor genes is found.
 oli. The lymphocytes are of post-thymic origin. The small       Common chromosome abnormalities in 80 % of T-PLL
 cell variant (T-CLL) may lack visible nucleoli. In some         patients are inversion of chromosome 14 with breakpoints in
 cases, the nuclei of this leukemia can be very irregularly      the long arm at q11 and q32. Frequent findings are abnor-
 formed and reveal cerebriform features. Usually, the periph-    malities of chromosome 8, abnormalities of chromosome 6
 eral blood, bone marrow and lymph nodes are infiltrated,        and 17 have also been described [69]. Some cases show dele-
 involvement of liver, spleen, and skin may occur. The bone      tion of the TP53 gene.
 marrow shows an interstitial or diffuse infiltration.
194                                                                                                                     7   Malignant Lymphomas
Case History (T-cell chronic lymphocytic leukemia                               The bone marrow biopsy showed diffuse fibrosis and
[T-CLL]/T-cell prolymphocytic leukemia [T-PLL])                             slight to moderate hypercellularity. An interstitial and diffuse
A 65-year-old woman was treated with cortisone for acute hearing            infiltration by small lymphocytes, with an identical immuno-
loss and iridocyclitis. The patient developed leucocytosis, general-        phenotype to that described in the lymph node, was seen (see
ized lymphadenopathy and splenomegaly. A cervical lymph node                Fig. 7.33). The amount of the T-cell infiltration, in relation to
extirpation and bone marrow biopsy were performed.                          the still normoquantitative hematopoiesis was 25%.
                                                                                Diagnosis was infiltration of lymph node and bone mar-
    The lymph node was diffusely infiltrated with some unaf-                row by T-PLL.
fected follicles still present (see Figs. 7.31 and 7.32). The                   Because of the rarity of this case, in addition to T-cell
infiltrating cells were small lymphoid cells with round nuclei              receptor gene analysis, which showed clonally rearranged
without visible nucleoli. These cells were positive with anti-              TCR beta and TCR gamma chain genes, low density whole
bodies to CD2, CD3, CD4, CD5, CD7, CD52 (very strong                        genome copy number variation (CNV) analysis using Ion
positivity), and BCL2 and negative for antibodies to CD8,                   Torrent NGS (Next-Generation Sequencing) was performed
CD16, CD57, TIA1, and granzyme B. The proliferation frac-                   (see Fig. 7.32c).
tion (MIB1) was very low with less than 5%
a b
c d
CD3 CD4
Fig. 7.31 T-PLL (lymph node). (a, b) Diffuse infiltration of the lymph      and CD7-positive (right side). (g) No CD8 reactivity is seen in the infil-
node by small lymphocytes sparing follicles (H&E). (c) Infiltrating         trating lymphocytes. (h) Lymphocytes are strongly stained with an anti-
lymphocytes are CD3-positive. (d) Strong CD4-positivity of infiltrating     body to CD52
lymphocytes. (e, f) Infiltrating lymphocytes are CD5-positive (left side)
7       Malignant Lymphomas                                                                                                           195
e f
CD5 CD7
g h
CD8 CD52
a b
CD20
Fig. 7.32 T-PLL lymph node. (a) Preserved follicles are highlighted   (c) CNV-analysis using next-generation sequencing shows deletion 9p,
with an antibody to CD20. (b) Low proliferation fraction within       deletion 9q12-22, deletion 11q14-24
the neoplastic cell population as shown with an antibody to MIB1.
196                                                                                                                       7   Malignant Lymphomas
a b
c d
CD3 CD4
Fig. 7.33 T-PLL (bone marrow). (a, b) Hypercellular bone marrow                and CD8-negative (right side). (g) Strong reactivity of neoplastic lym-
diffusely infiltrated by small lymphocytes (H&E). (c, d) Diffuse infil-        phocytes with an antibody to CD52. (h) Slight fibrosis can be seen
tration of the bone marrow by CD3 (left side) and CD4 (right side)-            between the infiltrating lymphocytes (Gomori’s stain)
positive lymphocytes. (e, f) Infiltrating cells are CD7-positive (left side)
7   Malignant Lymphomas                                                                                                197
e f
CD7 CD8
g h
CD52
a b
c d
CD3
Fig. 7.34 Case 1. (a, b) Slightly hypercellular bone marrow (H&E)        (f) CD8-positive lymphocytes are markedly increased. (g) There is an
with a slight increase of lymphocytes and a small lymphocytic aggre-     increase of CD57-  positive lymphocytes which are predominantly
gate. (c) Slight reticulin fibrosis is revealed by Gomori’s stain. (d)   located intrasinusoidal. (h) Cytotoxic phenotype is revealed by using
Interstitial increase of small- to medium-sized CD3-positive             granzyme B
T-lymphocytes. (e) Compared to CD3, there is a loss of CD7-positivity.
200                                7   Malignant Lymphomas
e f
CD7 CD8
g h
CD57 granzymeB
a b
c d
CD20 CD3
Fig. 7.35 Case 2. (a, b) In H&E stained sections, hypercellular bone        ing CD3 (d) and CD8 (e). (f) Only a few CD5-positive lymphocytes can
marrow is seen. The higher magnification (b) reveals dysplastic hema-       be seen corresponding to a loss of T-cell marker. (g) Typically, the
topoiesis with prominent megaloblastic erythropoiesis. (c) A reactive       T-cell population shows reactivity for granzyme B. (h) Increase of
lymphoid aggregate is mainly composed of CD20-positive                      CD34-positive blasts according to myelodysplasia with excess of
B-lymphocytes. (d, e) Interstitial infiltration of T-lymphocytes express-   blasts-1
202                                                                                               7   Malignant Lymphomas
e f
CD8 CD5
g h
granzymeB CD34
7.15     Hepatosplenic T-Cell Lymphoma                         which is mostly involved, reveals a marked intrasinusoidal
          (HSTL)                                                infiltration pattern (Fig. 7.37). Spleen and liver are enlarged;
                                                                splenic infiltration occurs in the red pulp (Fig. 7.36), the liver
7.15.1 Epidemiology                                            shows intrasinusoidal infiltration.
Case History (HSTL)                                                       The massively enlarged spleen showed multiple infarc-
A 35-year-old male with Crohn’s disease had been treated                  tions and an infiltration of the red pulp by neoplastic
with Imurek® for more than 6 years. He presented with                     lymphocytes with the immunophenotype of HSTL
weight loss, fatigue, pancytopenia, and massive hepato-                   (Fig. 7.36). The bone marrow was infiltrated by the same
splenomegaly as well as purpura. Splenectomy, because                     tumor cell population interstitial and intrasinusoidal
of infarction and a bone marrow biopsy were performed.                    (Fig. 7.37).
a b
c d
CD3
e f
CD4 CD5
Fig. 7.36 HSTL. (a) Overview shows marked expansion of splenic red        CD3-positivity of the infiltrating cells with sparing of focally visible
pulp and atrophy of follicles. (b, c) Higher magnification reveals lym-   white pulp. (e–g) The neoplastic cells do not react with antibodies to CD4
phatic infiltration of the red pulp by monomorphic medium-sized lym-      (e), CD5 (f), and CD7 (g). (h) Only a few lymphocytes are stained with
phocytes with small nucleoli and a rather pale cytoplasm. (d) Strong      an antibody to CD8. Note the sinusoidal lining by CD8-positive cells
7   Malignant Lymphomas                                                                                                                            205
g h
CD7 CD8
a b
c d
Fig. 7.37 HSTL in the bone marrow. (a–d) Hypercellular bone mar-            of intrasinusoidal lymphocytes by CD2. (f, g) CD3-positive neoplastic
row biopsy with interstitial and intrasinusoidal infiltration by atypical   cells infiltrate the bone marrow in an interstitial and intrasinusoidal pat-
lymphatic cells. Especially in the highest magnification (d) sinusoidal     tern. (h) Tumor cells are TIA1-positive
infiltration by monomorphic lymphocytes is visible. (e) Weak staining
206                                                                                                7   Malignant Lymphomas
e f
CD2 CD3
g h
CD3 TIA1
Case History (MF/SS) (see Fig. 7.38)                                       biopsy showed a diffuse and focally nodular infiltration by
A 59-year-old female patient suffering from generalized                    neoplastic T-lymphocytes with many intermingled CD30-
skin disease with erythroderma was treated for MF for 2                    positive blasts (Fig. 7.38). A transformation of SS could be
years. Because the patient did not respond to treatment, a                 diagnosed with a percentage of 70% in relation to
bone marrow biopsy was performed. This bone marrow                         hematopoiesis.
a b
c d
CD3 CD4
Fig. 7.38 Case history MF/SS. (a, b) The bone marrow is hypercellu-        SS-cells showing positivity for CD3 (c), CD4 (d), and CD5 (e). (f) Loss
lar due to a neoplastic lymphocytic infiltration (a) with many intermin-   of the T-cell marker CD7. (g) Only a few reactive small CD8-positive
gled blasts (b). (c–e) The infiltrating cells reveal the phenotype of      lymphocytes can be detected. (h) Many blasts with CD30-positivity
7   Malignant Lymphomas                                                                                             209
e f
CD5 CD7
g h
CD8 CD30
7.17.1 Epidemiology
                                                                 7.17.3 Immunohistochemistry
PTCLs, NOS account for 10–15% of lymphoproliferative
disorders in Western countries [79]. Mainly, this lymphoma       PTCL, NOS in most cases show an aberrant T-cell phenotype
occurs in adults and is extremely rare in children. There is a   with frequent downregulation or loss of CD5 and CD7. In nodal
male: female ratio of 2.1.                                       cases, CD4+ and CD8− cases predominate, but double positiv-
                                                                 ity or double negativity can be detected in some cases.
                                                                 Expression of CD30, CD56, and cytotoxic markers is variable.
7.17.2 Morphology                                               More than half of the cases are CD52-negative. Aberrant expres-
                                                                 sion of one or more B-cell markers (mainly CD20 and CD79a)
The morphological appearance of PTCL, NOS is quite               is seen in a few cases. The proliferation fraction, detected with
variable from monomorphous to highly polymorphous                MIB1 is usually high. EBV-positivity is not uncommon.
infiltration. In most cases, medium-sized and large lym-
 phocytes with irregular formed nuclei, prominent nucle-
 oli, and many mitoses can be seen. Cells with a rather          7.17.4 Cytogenetic Abnormalities
 clear cytoplasm and Reed-Sternberg (RS-)-like cells may                 and Molecular Characteristics
 be present. But there are also cases with mainly small
 lymphoid cells with irregular nuclei. The frequency of          In most cases, clonal rearrangement of TCR is found. PTCLs,
 bone marrow infiltration varies greatly from study to           NOS often show complex karyotypes [82]. The genetic
 study; on average one-third of PTCL, NOS show bone              imbalances detected in PTCL, NOS vary from those of other
 marrow infiltration [80]. The infiltration pattern described    T-cell lymphomas.
7   Malignant Lymphomas                                                                                                                   211
Case History (PTCL, NOS) (see Fig. 7.39)                                 was moderately hypercellular with increase of all hematopoietic
A 39-year-old female presented with hemolytic anemia and                 cells. In addition, a diffuse and multifocal patchy infiltration by
generalized lymphadenopathy. In a cervical lymph node infiltra-          PTCL, NOS was seen (Fig. 7.39). The percentage of the infiltra-
tion by PTCL, NOS was diagnosed. The bone marrow biopsy                  tion in relation to hyperplastic hematopoiesis was 20%.
a b
c d
GlycC MPO
Fig. 7.39 PTCL, NOS. (a, b) H&E stained section shows hypercellu-        (d) Slightly increased myelopoiesis is revealed by an antibody to
lar bone marrow with hyperplastic erythropoietic cells and increase of   myeloperoxidase. (e, f) Diffuse infiltration and several small patchy
lymphatic cells. (c) Erythropoiesis is markedly increased (consistent    aggregates of CD3-positive lymphocytic cells. (g, h) The lymphoma
with hemolytic anemia) as shown with an antibody to glycophorin C.       cells show double positivity for CD4 (g) and CD8 (h)
212                                                                                             7   Malignant Lymphomas
e f
CD3 CD3
g h
CD4 CD8
Case History (AITL) (Fig. 7.40)                                           diagnosed as infiltration by AITL. The hypercellular bone
A 74-year-old woman was admitted to the hospital with gen-                marrow biopsy showed multiple nodular to patchy mostly
eralized lymphadenopathy and weight loss of 10 kg within                  intertrabecular infiltrates consistent with an infiltration by
the last 4 months. The biopsy of a cervical lymph node was                AITL with an amount of neoplastic infiltration of 30%.
a b
c d
CD3 CD4
Fig. 7.40 AITL. (a) In routinely H&E stained sections, the bone mar-      positivity with an antibody to CD3. (d) There is a high amount of
row is markedly hypercellular due to multiple atypical nodular to         CD4-positive tumor cells. (e) CXCL13 is expressed by most neoplas-
patchy intertrabecular infiltrates. (b) Higher magnification reveals an   tic cells. (f) Intermingled, a few reactive CD8-positive T-cells are pres-
infiltrate composed of medium-sized lymphocytes, plasma cells, and        ent. (g) CD30-positive blasts can be detected within the infiltrates.
blasts (H&E). (c) Most cells within the infiltrate are T-cells showing    (h) Some EBV-positive cells are visualized by EBER in situ hybridization
7   Malignant Lymphomas                                                                                                     215
e f
CXCL13 CD8
g h
CD30 EBER
7.18.5 Caution                                                  classical Hodgkin lymphoma and even diffuse large B-cell
                                                                 lymphoma, especially if CD20 and CD30-positive EBV-
In contrast to AITL in the lymph node, CD10 is rarely            positive cells are found. In such cases, a descriptive diagno-
expressed by the tumor cells in the bone marrow.                 sis should be made (e.g., bone marrow infiltration by
   A proliferation of follicular dendritic cells (FDC), a com-   malignant lymphoma with many T-cells and intermingled
mon feature in the lymph node, is scarcely seen in infiltrates   B-blasts: differential diagnosis comprises AITL, classical
of the bone marrow.                                              Hodgkin lymphoma and DLBCL). PCR-studies for clonality
   If the bone marrow biopsy is taken without a previous         are essential in such cases to classify the lymphoma
lymph node, infiltrates of AITL may resemble infiltrates of      correctly.
216                                                                                                  7   Malignant Lymphomas
In the lymph node, ALCL shows a variable morphologic            About 90% of ALCL show clonal rearrangement of the
spectrum. The hallmark cells are large lymphoid cells           T-cell receptor genes. The most frequent genetic abnormal-
with an often horseshoe-shaped nucleus. The cytoplasm           ity is a translocation t(2;5)(p23;q35) between the ALK
of the tumor cells is mostly abundant. An infiltration of       gene on chromosome 2 and the nucleophosmin gene on
the bone marrow is rather rare with about 25% [85]. The         chromosome 5. There are variant translocations between
infiltration pattern is either interstitial and/or vaguely      ALK and other genes on chromosomes 1, 2, 3, 17, 19, 22,
nodular or intrasinusoidal. If hallmark cells are present,      and X. Using RT-PCR, the t(2;5) can be detected, but cases
these cells can easily be detected. In many cases, the infil-   with variant translocations are negative by standard RT-PCR
trate is subtle and is missed in routinely stained H&E sec-     method.
7   Malignant Lymphomas                                                                                                                        217
Case History (ALCL) (Figs. 7.41 and 7.42)                                    with an unknown primary tumor. An abdominal lymph
A 73-year-old male presented with weight loss, enlarged                      node (Fig. 7.42) and bone biopsy of lumbar vertebra
abdominal lymph nodes and bone pain in the shoulder as                       revealed an infiltration by ALCL, ALK-negative. A bone
well as the vertebral column and lower extremities.                          marrow biopsy was performed showing a vaguely nodular
Skeletal scintigraphy revealed numerous osteodestructive                     to patchy paratrabecular infiltrate by ALCL, ALK-negative
lesions within the bone, suggestive of metastatic process                    (Fig. 7.41).
a b
c d
CD30 CD30
Fig. 7.41 ALCL in the bone marrow. (a, b) In routinely stained H&E           nized in the overview. (e) Whereas the tumor cells are strongly CD2-
sections, a normocellular bone marrow is seen. There is a single patchy      positive (f), no reactivity is seen using CD3 (f). (g, h) Rather weak
neoplastic infiltrate consisting of large lymphoid cells. (c, d) The tumor   positivity with an antibody to CD4 (g) and strong positivity with a an
cells in the infiltrate are strongly CD30-positive, a staining even recog-   antibody to CD25 (h)
218                                                                                                                    7   Malignant Lymphomas
e f
CD2 CD3
g h
CD4 CD25
a b
Fig. 7.42 ALCL in the lymph node. (a, b) To a large extent, the lymph        (f) Weak CD4-positivity is found in a few blasts. (g) Typically, the
node shows an intact architecture (a). In higher magnification (b), an       tumor cells of ALCL are BCL2-negative. (h) High proliferation fraction
accentuated intrasinusoidal infiltration by huge blasts is obvious. (c, d)   of the neoplastic cells (MIB1)
The intrasinusoidal blasts are strongly positive for CD30 and (e) EMA.
7   Malignant Lymphomas                                                                                          219
c d
CD30 CD30
e f
EMA CD4
g h
BCL2 MIB1
7.20     Adult T-Cell Leukemia/Lymphoma                          coarsely granular with distinct, occasionally prominent
          (ATLL)                                                  nucleoli. A variable amount of blast-like cells is usually pres-
                                                                  ent. The infiltration pattern of the bone marrow is usually
7.20.1 Epidemiology                                              patchy or diffuse to dense.
Case History (ATLL) (Fig. 7.43)                                              cells, which were medium sized with a variable amount of
A 42-year-old HIV-negative African male presented with                       intermingled larger blasts. A few eosinophils were intermin-
bone pain and dysphagia due to enlarged tonsils. CT scan                     gled, occasionally hematopoietic cells could be detected.
revealed masses of osteolytic lesions in the whole skeletal                  The immunohistochemistry revealed positivity of the tumor
system. Notable laboratory values at presentation revealed a                 cells with antibodies to CD2, CD3, CD4, CD5, CD99, and
white blood cell count of 32,000/L, hemoglobin of 11.8 g/                    FOXP3. The blasts showed variable reactivity with an anti-
dL, thrombocytopenia with 82,000/L, and a high LDH with                      body to CD30. Cytotoxic markers and CD7 were negative.
780 U/L. A bone marrow biopsy from an osteolytic lesion in                   HTLV-1 antibody testing was positive by enzyme-linked
the iliac crest was performed (Fig. 7.43). The bone marrow                   immunosorbent assay. The diagnosis of ATLL with more
was densely infiltrated by highly pleomorphic lymphatic                      than 80% infiltration of the bone marrow was confirmed.
a b
c d
CD2
Fig. 7.43 Case history ATLL. (a–c) In routinely stained H&E sections,        infiltrating tumor cells with an antibody to CD7, only a few small reac-
there is a dense bone marrow infiltration by highly pleomorphic lym-         tive T-lymphocytes are stained. The inset highlights a huge CD30-
phoid cells. Many mitoses are seen, occasional larger blasts with promi-     positive blast. (g) Strong reactivity of the neoplastic cells is seen with an
nent nucleoli are visible. The infiltrating pleomorphic cells are strongly   antibody to CD25. (h) Intranuclear reactivity is seen in most tumor cells
CD2-positive (d) and CD4-positive (e). (f) There is no reactivity of the     using an antibody to FOXP3
222                                7   Malignant Lymphomas
e f
CD30
CD4 CD7
g h
CD25 FOXP3
7.21     Hodgkin Lymphoma                                         cells [91]. Within the bone marrow infiltrates small lympho-
                                                                   cytes, scattered LP-cells and plasma cells as well as histio-
7.21.1 N
        odular Lymphocyte Predominant                             cytes are present. Some cases with NLPHD show a follicular
       Hodgkin Lymphoma (NLPHD)                                    pattern of infiltration [92].
a b
c d
CD20 CD21
Fig. 7.44 NLPHD. (a, b) Routinely stained H&E sections of the            The LP-cells are characteristically J-chain-positive. (f) Despite
lymph node show a vaguely nodular neoplastic infiltrate (a) consisting   fragmentation, a nodular and patchy neoplastic infiltrate is visible
of small lymphocytes with scattered LP-cells (b). (c) The LP-cells are   in the bone marrow biopsy. (g, h) MGG-stain and H&E stain reveal
highlighted with an antibody to CD20; also, small lymphocytes are        an infiltrate with small lymphocytes, histiocytes, and huge blasts
CD20-positive B-cells. (d) Within the nodules an irregular network       consistent with LP-cells
of follicular dendritic cells is stained with an antibody to CD21. (e)
7   Malignant Lymphomas                                                                                             225
e f
J-chain MGG
g h
MGG
7.22     Classical Hodgkin Lymphoma (cHL)                            focal bone marrow infiltration with nodular or patchy infiltrates
                                                                      surrounded by normal bone marrow. A minority of patients
7.22.1 Epidemiology                                                  with cHL reveal a paratrabecular infiltration. The infiltrates are
                                                                      polymorphous with small lymphocytes, histiocytes, and plasma
High variability is reported on the incidence of BM infiltra-         cells as well as eosinophils. The infiltrates are accompanied by
tion by cHL. In children, the incidence is very low with              a reticulin or collagen fibrosis confined to the lesions.
reported 1.8% in a large series [93], thus the significance of
a bone marrow biopsy is questionable in pediatric patients.
In adults, the reported incidence of bone marrow involve-             7.22.3 Immunohistochemistry
ment is 2–32%. In mixed cellularity Hodgkin lymphoma
bone marrow involvement is more frequent than in nodular              HRS show a typical immunohistochemical profile, facilitat-
sclerosis type [94].                                                  ing the detection in bone marrow biopsies. The tumor cells
   Infection by EBV may play a role in the pathogenesis of            are always CD30-positive (Fig. 7.45e), often reactive with
cHL. While in endemic areas nearly 100% of Hodgkin lym-               antibodies to CD15, CD20, CD23, PAX5, BCL6, and MUM1.
phomas are EBV-positive, in Western countries EBV-                   Usually, the tumor cells do not express LCA and BCL2.
positivity is found only in a proportion of cases [95].                   The tumor cells, especially of mixed cellularity type often
                                                                      express EBV-associated markers, such as LMP1 and are pos-
                                                                      itive for Epstein–Barr encoding region (EBER) in situ
7.22.2 Morphology (Figs. 7.45 and 7.46)                              hybridization (Fig. 7.45h) [96].
a b
c d
GOM CD20
Fig. 7.45 cHL case 1. (a, b) Routinely stained H&E section shows            The tumor cells are strongly CD20-positive. (e) Specific CD30 staining
hypercellular bone marrow with multiple patchy infiltrates (a), which,      of the tumor cells. (f) Some tumor cells are CD15-positive; further-
in higher magnification (b) reveal HRS-cells in a background of poly-       more, the intermingled eosinophils are CD15-positive. (g) The small
morphic lymphocytes, histiocytes, and granulocytes. (c) Within the          polymorphic lymphocytes are T-cells (CD3). (h) Tumor cells are EBV-
neoplastic infiltrates, a dense fibrosis is evident (Gomori’s stain). (d)   positive (EBER)
228                                7   Malignant Lymphomas
e f
CD30 CD15
g h
CD3 EBER
a b
c d
Fig. 7.46 cHL, case 2. (a–b) Bone marrow biopsy is densely infil-             cytes are strongly CD20-positive. (f) Granulocytes and some of the
trated by a neoplastic infiltrate (a). In higher magnification (c–d) atypi-   mononuclear blasts are CD15-positive. (g) Strong CD30-positivity of
cal, mainly mononuclear blasts can be identified in an inflammatory           the neoplastic cells (h) Typically, HRS-cells are stained for the
background. (e) The mononuclear blasts as well as some small lympho-          transferrin-receptor CD71, an activation antigen
230                                7   Malignant Lymphomas
e f
CD20 CD15
g h
CD30 CD71
7.22.5 Caution                                                         11. Krause JR, Drinkard LC, Keglovits LC. Hodgkin lymphoma trans-
                                                                            formation of chronic lymphocytic leukemia/small lymphocytic
                                                                            lymphoma. Proc (Bayl Univ Med Cent). 2013;26(1):16–8.
Several reactive and neoplastic diseases may mimic BM 12. Schroers R, Griesinger F, Trümper L, Haase D, Kulle B, Klein-
infiltration by cHL. Reactive disorders are, for example,                   Hitpass L, Sellmann L, Dührsen U, Dürig J. Combined analysis
granulomatous and histiocyte-rich lesions. Neoplastic disor-                of ZAP-70 and CD38 expression as a predictor of disease pro-
                                                                            gression in B-cell chronic lymphocytic leukemia. Leukemia.
ders are ALCL, Richter’s syndrome in CLL-patients, T-cell-                 2005;19(5):750–8.
rich, and histiocyte-rich B-cell lymphoma, AITL, systemic 13. Crespo M, Bosch F, Villamor N, et al. ZAP-70 expression as a sur-
mast cell disease, and metastatic carcinoma. Moreover,                      rogate for immunoglobulin-variable-region mutations in chronic
EBV-associated lymphoproliferative disorders in primary or                  lymphocytic leukemia. N Engl J Med. 2003;348:1764–75.
                                                                        14. Moreno C, Montserrat E. Genetic lesions in chronic lympho-
secondary immunodeficiency may resemble cHL. In most                        cytic leukemia: what’s ready for prime time use? Haematologica.
instances, these diseases can be discriminated by appropriate               2010;95(1):12–5.
immunohistochemical and molecular studies.                              15. Admirand JH, Knoblock RJ, Coombes KR, Tam C,
    Challenging differential diagnoses include other CD30-                 Schlette EJ, Keating MJ, Luthra R, Medeiros LJ, Abruzzo
                                                                            LV. Immunohistochemical detection of ZAP70 in chronic lympho-
positive neoplasms such as peripheral T-cell lymphomas                      cytic leukemia predicts immunoglobulin heavy chain gene mutation
NOS, ALCL, EBV-positive DLBCL, and rare cases of                            status and time to progression. Mod Pathol. 2010;23(11):1518–23.
CD30-positive mast cell neoplasms. However, cHL infiltra- 16. Randen U, Tierens AM, Tjønnfjord GE, Delabie J. Bone marrow
tion is always accompanied by a characteristic stromal                      histology in monoclonal B-cell lymphocytosis shows various B-cell
                                                                            infiltration patterns. Am J Clin Pathol. 2013;139(3):390–5.
change with inflammatory cells and fibrosis.                            17. Landgren O, Albitar M, Ma W, Abbasi F, Hayes RB, Ghia P, Marti
    Not involved areas of the bone marrow in patients with                  GE, Caporaso NE. B-cell clones as early markers for chronic lym-
cHL often show nonspecific alterations. Such changes are                    phocytic leukemia. N Engl J Med. 2009;360(7):659–67.
eosinophilia, myeloid hyperplasia, edema, reactive plasma-              18. Demir HA, Bayhan T, Üner A, Kurtulan O, Karakuş E, Emir S,
                                                                            Özyörük D, Ceylaner S. Chronic lymphocytic leukemia in a child: a
cytosis, granulomas, megakaryocytic hyperplasia, or                         challenging diagnosis in pediatric oncology practice. Pediatr Blood
increased bone remodeling and fibrosis. The most common                     Cancer. 2014;61(5):933–5. https://doi.org/10.1002/pbc.24865.
finding is myeloid hyperplasia.                                             Epub 2013 Nov 19.
                                                                        19. Galton DA, Goldman JM, Wiltshaw E, et al. Prolymphocytic leuke-
                                                                            mia. Br J Haematol. 1974;27:7–23.
                                                                        20. Orazi O, Foucar K, Knowles DM, Weiss LM, editors. Knowles’
References                                                                  neoplastic pathology. 3rd ed. Philadelphia: Wolters Kluwer/
                                                                            Lippincot William & Wilkins; 2014. p. 434–5.
  1. Swerdlow SH, Campo E, Harris NL, Jaffe ES, Pileri SA, Stein H, 21. Lens D, Matutes E, Catovsky D, Coignet LJA. Frequent deletions
     Thiele J, Vardiman JW, editors. WHO classification of tumours of       at 11q23 and 13q14 in B cell prolymphocytic leukemia (B.PLL).
     haematopoietic and lymphoid tissues. Lyon: IARC; 2017.                 Leukemia. 2000;14:427–43.
  2. Swerdlow SH, et al. Mantle cell lymphoma. In: Swerdlow SH, 22. Flatley E, Chen A, Jaffe ES, Dunlap JB, Pittaluga S, Abdullah S,
     et al., editors. WHO classification of tumours of haematopoietic       Olson SB, Spurgeon SE, Fan G. Aberrations of MYC are a com-
     and lymphoid tissues. Lyon: IARC Press; 2017. p. 285–90.               mon event in B-cell prolymphocytic leukemia. Am J Clin Pathol.
  3. Swerdlow SH, Campo E, Harris NL, Jaffe ES, Pileri SA, Stein H,         2014;142(3):347–54.
     Thiele J, Vardiman JW, editors. WHO classification of tumours of 23. Phekoo KJ, Jack RH, Davies E, et al. The incidence and survival
     haematopoietic and lymphoid tissues. Lyon: IARC Press; 2017.           of Waldenstrom’s macroglobinaemia in South East England. Leuk
  4. Arber DA. Molecular diagnostic approach to Non-Hodgkin’s               Res. 2008;32(1):55–9.
     Lymphoma. J Mol Diagn. 2000;2(4):178–90.                           24. Merlinig G, Baldini L, Broglia C, et al. Prognostic factors in
  5. Qi Q, Liu Y, Cheng Y, Glanville J, Zhang D, Lee J-Y, Olshen RA,        symptomatic Waldenstrom’s macroglobuinemia. Semin Oncol.
     Weyand CM, Boyd SD, Goronzy JJ. Diversity and clonal selec-            2003;30(2):211–5.
     tion in the human T-cell repertoire. Proc Natl Acad Sci USA. 25. Kristinsson SY, Bjorkholmm M, Goldin LR, et al. Risk of lym-
     2014;111(36):13139–44.                                                 phoproliferative disorders among first-degree relatives of lympho-
  6. De Lima M, O’Brien S, Lerner S, Keating ML. Chronic lympho-            plasmacytic lymphoma/Waldenstrom macroglobuliaemia patients:
     cytic leukemia in the young patient. Semin Oncol. 1998;25:107–16.      a population based study in Sweden. Blood. 2008;112(8):3052–6.
  7. Parikh SA, Neil E, Kay NE, Shanafelt TD. Monoclonal B-cell lym- 26. Treon SP, Hunter ZR, Aggarwal A, et al. Characterization
     phocytosis: update on diagnosis, clinical outcome, and counseling.     of familial Waldenstrom’s macroglobulinemia. Ann Oncol.
     Clin Adv Hematol Oncol. 2013;11(11):720–9.                             2006;17(3):488–94.
  8. Balatti V, Bottoni A, Palamarchuk A, Alder H, Rassenti LZ, Kipps 27. van de Donk NWCJ, Palumbo A, Johnsen HE, Engelhardt M, Gay
     TJ, Pekarsky Y, Croce CM. NOTCH1 mutations in CLL associated           F, Gregersen H, Hajek R, Kleber M, Ludwig H, Morgan G, Musto
     with trisomy 12. Blood. 2012;119(2):329–31.                            P, Plesner T, Sezer O, Terpos E, Waage A, Zweegman S, Einsele
  9. Geisler C, Ralfkiaer E, Hansen MM, Hou-Jensen K, So L. The             H, Sonneveld P, Lokhorst HM. The clinical relevance and manage-
     bone marrow histological pattern has independent prognostic value      ment of monoclonal gammopathy of undetermined significance and
     in early stage chronic lymphocytic leukaemia. Br J Haematol.           related disorders: recommendations from the European Myeloma
     1986;62:47–54.                                                         Network. Haematologica. 2014;99(6):984–96.
10. Jahic A, Iljazovic E, Arnautovic-Custovic A, Halilbasic A, Simendic 28. Morice WG, Chen D, Kurtin PJ, et al. Novel immunopheno-
     V, Zabic A. Prognostic significance of bone-marrow pattern and         typic features of marrow lymphoplasmacytic lymphoma and cor-
     immunophenotypic score in B-chronic lymphocytic leukemia at            relation with Waldenstrom’s macroglobulinemia. Mod Pathol.
     diagnosis. Med Arch. 2011;65(3):132–6.                                 2009;22(6):807–16.
232                                                                                                                  7   Malignant Lymphomas
29. Rossi D. Role of MYD88 in lymphoplasmacytic lymphoma diagno-           49. Pich A, Fraire F, Fornari A, Bonino LD, Godio L, Chiusa L,
    sis and pathogenesis. Hematology Am Soc Hematol Educ Program.              Palestro G. Intrasinusoidal bone marrow infiltration and splenic
    2014;2014(1):113–8.                                                        marginal zone lymphoma: a quantitative study. Eur J Haematol.
30. Sargent RL, Cook JR, Aguilera NI, et al. Fluorescent immunopheno-          2006;76(5):392–8.
    typic and interphase cytogenetic characterization of nodal lympho-     50. Mateo M, Mollejo M, Villuendas R, Algara P, Sanchez-Beato M,
    plasmacytic lymphoma. Am J Surg Pathol. 2008;32(11):1643–53.               Martinez P, Piris MA. 7q31-32allelic loss is a frequent finding in
31. Smedby KE, Hjalgrim H. Epidemiology and etiology of mantle                 splenic marginal zone lymphoma. Am J Pathol. 1999;154:1583–9.
    cell lymphoma and other non-Hodgkin lymphoma subtypes. Semin           51. Foucar K. Bone marrow pathology. 2nd ed. Chicago: ASCP; 2001.
    Cancer Biol. 2011;21(5):293–8.                                             p. 453.
32. Cohen PL, Kurtin PJ, Donovan KA, Hanson CA. Bone marrow                52. Costes V, Duchayne E, Taib J, Delfour C, Rousset T, Baldet P,
    and peripheral blood involvement in mantle cell lymphoma. Br J             Delsol G, Brousset P. Intrasinusoidal bone marrow infiltration: a
    Haematol. 1998;101(2):302–10.                                              common growth pattern for different lymphoma subtypes. Br J
33. Schenka AA, Gascoyne RD, Duchayne E, Delsol G, Brousset                    Haematol. 2002;119(4):916–22.
    P. Prominent intrasinusoidal infiltration of the bone marrow by        53. Zullo A, Hassan C, Ridola L, Repici A, Manta R, Andriani
    mantle cell lymphoma. Hum Pathol. 2003;34(8):789–91.                       A. Gastric MALT lymphoma: old and new insights. Ann
34. Zeng W, Fu K, Quintanilla-Fend L, et al. Cyclin D1-negative blas-          Gastroenterol. 2014;27(1):27–33.
    toid mantle cell lymphoma identified by Sox11 expression. Am J         54. Bacon CM, Du M-Q, Dogan A. Mucosa-associated lymphoid tis-
    Surg Pathol. 2012;36(2):214–9.                                             sue (MALT) lymphoma: a practical guide for pathologists. J Clin
35. Stefancikova L, et al. Loss of the p53 tumor suppressor activity is        Pathol. 2007;60(4):361–72.
    associated with negative prognosis of mantle cell lymphoma. Int J      55. Inamdar KV, Medeiros LJ, Jorgenen JL, Amin HM, Schlette
    Oncol. 2010;36(3):699–706.                                                 EJ. Bone marrow involvement by marginal zone B cell lymphomas
36. Burke JS. The value of the bone-marrow biopsy in the diagnosis of          of different types. Am J Clin Pathol. 2008;129:714–22.
    hairy cell leukemia. Am J Clin Pathol. 1978;70:876–84.                 56. Kajiura D, Yamashita Y, Mori N. Diffuse large B cell lymphoma
37. Golomb HM, Catovsky D, Golde DW. Hairy cell leukemia: a clini-             initially manifesting in the bone marrow. Am J Clin Pathol.
    cal review based on 71 cases. Ann Intern Med. 1978;89:677.                 2007;127(5):762–9.
38. Sherman MJ, Hanson CA, Hoyer JD. An assessment of the useful-          57. Chung R, Lai R, Wei P, et al. Concordant but not discordant bone
    ness of immunohistochemical stains in the diagnosis of hairy cell          marrow involvement in diffuse large B cell lymphoma predicts a
    leukemia. Am J Clin Pathol. 2011;136(3):390–9.                             poor clinical outcome independent of the International Prognostic
39. Cawley JC. The pathophysiology of the hairy cell. Hematol Oncol            Index. Blood. 2007;110(4):1278–82.
    Clin North Am. 2006;20:1011–21.                                        58. Campbell J, Seymour JF, Matthews F, et al. The prognostic
40. Tiacci E, Schiavoni G, Forconi F, Santi A, Trentin L, Ambrosetti           impact of bone marrow involvement in patients with diffuse large
    A, Cecchini D, Sozzi E, Francia di Celle P, Di Bello C, Pulsoni A,         cell lymphoma varies according to the degree of infiltration and
    Foà R, Inghirami G, Falini B. Simple genetic diagnosis of hairy            presence of discordant marrow involvement. Eur J Haematol.
    cell leukemia by sensitive detection of the BRAF-V600E mutation.           2006;76(6):473–80.
    Blood. 2012;119(1):192–5.                                              59. Choi WW, Weisenburger DD, Greiner TC, Piris MA, Banham
41. Falini B, Tiacci E, Liso A, Basso K, Sabattini E, Pacini R, Foa R,         AH, Delabie J, Braziel RM, Geng H, Iqbal J, Lenz G, Vose JM,
    Pulsoni A, Dalla FR, Pileri S. Simple diagnostic assay for hairy           Hans CP, Fu K, Smith LM, Li M, Liu Z, Gascoyne RD, Rosenwald
    cell leukaemia by immunocytochemical detection of annexin A1               A, Ott G, Rimsza LM, Campo E, Jaffe ES, Jaye DL, Staudt LM,
    (ANXA1). Lancet. 2004;363:1869–70.                                         Chan WC. A new immunostain algorithm classifies diffuse large
42. Whalin BE, Yri OE, Kimby E, Holte H, Delabie J, Smeland EB,                B-cell lymphoma into molecular subtypes with high accuracy. Clin
    Sundström C, Christensson B, Sander B. Clinical significance of            Cancer Res. 2009;15(17):5494–502.
    the WHO grades of follicular lymphoma in a population-based            60. Green TM, Young KH, Visco C, Xu-Monette ZY, Orazi A, Go
    cohort of 505 patients with long follow-up times. Br J Haematol.           RS, Nielsen O, Gadeberg OV, Mourits-Andersen T, Frederiksen
    2012;156(2):225–33.                                                        M, Pedersen M, Möller MB. Immunohistochemical double-hit
43. Coffing BN, Lim MS. Signet ring cell lymphoma in a patient with            score is a strong predictor of outcome in patients with diffuse
    elevated CA-125. J Clin Pathol. 2011;29:416–8.                             large B-cell lymphoma treated with rituximab plus cyclophospha-
44. Torlakovic E, Torlakovic G, Brunning RD. Follicular pattern of             mide, doxorubicin, vincristine, and prednisolone. J Clin Oncol.
    bone marrow involvement by follicular lymphoma. Am J Clin                  2012;30(28):3460–7.
    Pathol. 2002;118:780–6.                                                61. Bea S, Zettl A, Wright G, Salaverria I, Jehn P, Moreno V, Burek
45. Lossos IS, Alizadeh AA, Diehn M, Warnke R, Thorstenson Y,                  C, Ott G, Puig X, Yang L, Lopes-Guillermo A, Chan WC, Greiner
    Oefner PJ, Brown PO, Botstein D, Levy R. Transformation of                 TC, Weissenburger DD, Armitage JO, Gascoyne RD, Connors JM,
    follicular lymphoma to diffuse large-cell lymphoma: alternative            Grogan TM, Braziel R, Fisher RI, Smeland EB, Kvaloy S, Holte
    patterns with increased or decreased expression of c-myc and its           H, Delabie J, Simon R, Powell J, Wilson WH, Jaffe ES, Montserrat
    regulated genes. Proc Natl Acad Sci U S A. 2002;99:8886–91.                E, Müller-Hermelink HK, Staudt LM, Campo E, Rosenwald
46. Pittaluga S, Verhoef G, Criel A, et al. “Small” B-cell non-Hodgkin’s       A. Diffuse large B-cell lymphoma subgroups have distinct genetic
    lymphomas with splenomegaly at presentation are either mantle              profiles that influence tumor biology and improve gene-expression-
    cell lymphoma or marginal zone cell lymphoma: a study based on             based survival prediction. Blood. 2005;106:3183–90.
    histology, cytology, immunohistochemistry, and cytogenetic analy-      62. Dufau JP, Tourneau A, Molina T, Le Houcq M, Claessens YE,
    sis. Am J Surg Pathol. 1996;20:211–23.                                     Rio B, Delmer A, Diebold J. Intravascular large B-cell lymphoma
47. Olteanu H, Fenske TS, Harrington AM, Szabo A, MS PH, Kroft                 with bone marrow involvement at presentation and haemophago-
    SH. CD23 expression in follicular lymphoma. Clinicopathologic              cytic syndrome: two Western cases in favour of a specific variant.
    correlations. Am J Clin Pathol. 2011;135:46–53. https://doi.               Histopathology. 2000;37:509–12.
    org/10.1309/AJCP27YWLIQRAJPW.                                          63. Ferreri AJ, Campo E, Ambrosetti A, Ilariucci F, Seymour JF,
48. Catovsky D, Matutes E. Splenic lymphoma with circulating villous           Willemze R, Arrigoni G, Rossi G, Dell’Oro S, Lestani M, Asioli
    lymphocytes/splenicmarginal-zone lymphoma. Semin Hematol.                  S, Pedrinis E, Ungari M, Motta T, Rossi R, Artusi T, Iuzzolino P,
    1999;36:148–54.                                                            Zucca E, Cavalli F, Ponzoni M. Anthracycline-based chemother-
7     Malignant Lymphomas                                                                                                                          233
      apy as primary treatment for intravascular lymphoma. Ann Oncol.          78. Mao X, Lillington DM, Czepulowski B, Russel-Jones R, Young
      2004;15:1215–21.                                                             BD, Whittaker S. Molecular cytogenetic characterization of Sezary
64.   Wright DH. Burkitt’s lymphoma: a review of the pathology, immu-              syndrome. Genes Chromosomes Cancer. 2003;36:250–60.
      nology and possible aetiological factors. In: Sommers SC, editor.        79. Dogan A, Morice WG. Bone marrow histopathology in peripheral
      Pathology annual. New York: Appleton Century-Crofts; 1971.                   T-cell Lymphomas. Br J Haematol. 2004;127:140–54.
      p. 337–63.                                                               80. Caulet S, Delmer A, Audouin J, Letourneau A, Bernadou A, Zittoun
65.   Hummel M, Bentink S, Berger H, Klapper W, Wessendorf                         R, Diebold J. Histopathological study of bone marrow biopsies in
      S, Barth TF, Bernd HW, Cogliatti SB, Dierlamm J, Feller                      30 cases of T-cell lymphoma with clinical, biological and survival
      AC, Hansmann ML, Haralambieva E, Harder L, Hasenclever                       correlations. Hematol Oncol. 1990;8:155–68.
      D, Kuhn M, Lenze D, Lichter P, Martin-Subero JI, Moller                  81. White DM, Smith AG, Whitehouse JMA, Smith JL. Peripheral T
      P, Müller-Hermelink HK, Ott G, Parwaresch RM, Pott C,                        cell lymphoma: value of bone marrow trephine immunophenotyp-
      Rosenwald A, Rosolowski M, Schwaenen C, Sturzenhofecker B,                   ing. J Clin Pathol. 1989;42:403–8.
      Szczepanowski M, Trautmann H, Wacker HH, Spang R, Loeffler               82. Rizvi MA, Evens AM, Tallman MS, Nelson BP, Rosen ST. T-cell
      M, Trumper L, Stein H, Siebert R. A biologic definition of                   non-Hodgkin lymphoma. Blood. 2006;107:1255–64.
      Burkitt’s lymphoma from transcriptional and genomic profiling.           83. Grogg KL, Attygalle AD, Macon WR, Remstein ED, Kurtin PJ,
      N Engl J Med. 2006;354:2419–30.                                              Dogan A. Angioimmunoblastic T-cell lymphoma: a neoplasm of
66.   Salaverria I, Martin-Guerrero I, Wagener R, Kreuz M, Kohler                  germinal-center T-helper cells? Blood. 2005;106:1501–2.
      CW, Richter J, Pienkowska-Grela B, Adam P, Burkhardt B,                  84. Dogan A, Attygalle AD, Kyriakou C. Angioimmunoblastic T-cell
      Claviez A, Damm-Welk C, Drexler HG, Hummel M, Jaffe ES,                      lymphoma. Br J Haematol. 2003;121:681–91.
      Küppers R, Lefebvre C, Lisfeld J, Löffler M, Macleod RAF,                85. Fraga M, Brousset P, Schlaifer D, Payen C, Robert A, Rubie H,
      Nagel I, Oschlies I, Rosolowski M, Russell RB, Rymkiewicz G,                 Huguet-Rigal F, Delsol G. Bone marrow involvement in ana-
      Schindler D, Schlesner M, Scholtysik R, Schwaenen C, Spang R,                plastic large cell lymphoma. Immunohistochemical detection of
      Szczepanowski M, Trümper L, Vater I, Wessendorf S, Klapper W,                minimal disease and its prognostic significance. Am J Clin Pathol.
      Siebert R. A recurrent 11q aberration pattern characterizes a subset         1995;103:82–9.
      of MYC-negative high-grade B-cell lymphomas resembling Burkitt           86. Sadahira Y, Hata S, Sugihara T, Manabe T. Bone marrow involve-
      lymphoma. Blood. 2014;123:1187–98. https://doi.org/10.1182/                  ment in NPM-ALK-positive lymphoma: report of two cases. Pathol
      blood-2013-06-507996.                                                        Res Pract. 1999;195:657–61.
67.   Matutes E, Brito-Babapulle V, Swansbury J, Ellis J, Morilla              87. Stein H, Foss HD, Dürkop H, Marafioti T, Delsol G, Pulford K,
      R, Dearden C, Sempere A, Catovsky D. Clinical and labaro-                    Pileri S, Falini B. CD30(+) anaplastic large cell lymphoma: a
      tory features of 78 cases of T-prolymphocytic leukaemia. Blood.              review of its histopathologic, genetic, and clinical features. Blood.
      1991;78:3269–74.                                                             2000;96:3681–95.
68.   Dearden CE. T-cell prolymphocytic leukaemia. Med Oncol.                  88. Ohshima K, Jaffe ES, Kikuchi M. Adult T-cell leukemia/lymphoma.
      2006;23:17–22.                                                               In: Jaff ES, Harris NL, Stein H, Vardiman JW, editors. World Health
69.   Maljaei SH, Brito-Babapulle V, Hiorns LR, Catovsky                           Organization classification of tumours. Lyon: IARC Press; 2017.
      D. Abnormalities of chromosomes 8,11,14 and X in                             p. 363–7.
      T-prolymphocytic leukaemia studied by fluorescence in situ hybrid-       89. Graham RL, Burch M, Krause JR. Adult T-cell leukemia/lym-
      ization. Cancer Genet Cytogenet. 1998;103:110–6.                             phoma. Proc (Bayl Univ Med Cent). 2014;27(3):235–8.
70.   Robbi LG, Cooper B, Krause JR. T-cell prolymphocytic leukemia.           90. Khoury JD, Jones D, Yared MA, Manning JT Jr, Abruzzo LV,
      Proc (Bayl Univ Med Cent). 2013;26(1):19–21.                                 Hagemeister FB, Medeiros LJ. Bone marrow involvement in
71.   Morice WG, Kurtin PJ, Tefferi A, Hanson CA. Distinct bone mar-               patients with nodular lymphocyte predominant Hodgkin lym-
      row findings in T-cell granular lymphocytic leukemia revealed by             phoma. Am J Surg Pathol. 2004;28:489–95.
      paraffin section immunoperoxidase stains for CD8, TIA-1, and             91. Panjwani P, Epari S, Sengar M, Laskar S, Menon H, Shet T. Letter
      granzyme B. Blood. 2002;99(1):268–74.                                        to the Editor: Bone marrow involvement in nodular lymphocyte
72.   Morice WG, Jevremovic D, Hanson CA. The expression of the                    predominant Hodgkin lymphoma occurs in tumors with a variant
      novel cytotoxic proteine granzyme M by large granular lympho-                pattern. Leuk Lymphoma. 2015;56(1):236–8.
      cytic leukaemias of both T-cell and NK-cell lineage: an unexpected       92. Pillai G, Pezzella F, Gatter K. “Follicular” pattern of bone mar-
      finding with implications regarding the pathobiology of these dis-           row involvement in lymphocyte-predominant Hodgkin’s disease.
      orders. Br J Haematol. 2007;137:237–9.                                       Histopathology. 2003;43:203–5.
73.   Lamy T, Loughran TP. Current concepts: large granular lymphocyte         93. Mahoney DH Jr, Schreuders LC, Gresik MV, McClain KL. Role
      leukaemia. Blood Rev. 1999;13:230–40.                                        of staging bone marrow examination in children with Hodgkin dis-
74.   Narumi H, Kojima K, Matsuo Y, Shikata H, Sekiya K, Niya T,                   ease. Med Pediatr Oncol. 1989;30:175–7.
      Bando S, Niya H, Azuma T, Yakushijin Y, Sakai I, Yasukawa M,             94. Brunning RD, McKenna RW. Bone marrow lymphomas. In:
      Fujita S. T-cell large granular lymphocytic leukemia occurring               Brunning RD, McKenna RW, editors. Tumors of the bone marrow.
      after autologous peripheral blood stem cell transplantation. Bone            Atlas of tumor pathology. Third series, fascicle, vol. 9. Washington,
      Marrow Transplant. 2004;33:99–101.                                           DC: Armed Forces Institute of Pathology; 1994. p. 369–408.
75.   Rosh JR, Gross T, Mamula P, Griffiths A, Hyams J. Hepatosplenic          95. Leoncini L, Spina D, Nyongo A, Abinya O, Minacci C, Disanto
      T-cell lymphoma in adolescents and young adults with Crohn’s dis-            A, De Luca F, de Vivo A, Sabattini E, Poggi S, Pileri S, Tosi
      ease. A cautionery tale? Inflamm Bowel Dis. 2007;13:1024–30.                 P. Neoplastic cells of Hodgkin’s disease show differences in EBV
76.   Weidmann E. Hepatosplenic T cell lymphoma. A review on 45                    expression between Kenya and Italy. Int J Cancer. 1996;65:781–4.
      cases since the first report describing the disease as a distinct lym-   96. Pavlovic A, Durdov MG, Capkun V, Pitesa JJ, Sakic MB. Classical
      phoma entity in 1990. Leukaemia. 2000;14(6):991–7.                           Hodgkin lymphoma with positive Epstein-Barr virus status is
77.   Sibaud V, Beylot-Barry M, Thiébaut R, Parrens M, Vergier B,                  associated with more FOXP3 regulatory T cells. Med Sci Monit.
      Delaunay M, Beylot C, Chêne G, Ferrer J, de Mascarel A, Dubus                2016;22:2340–6.
      P, Merlio JP. Bone marrow histopathologic and molecular stag-            97. Ehlers A, Oker E, Benti nk S, Lenze D, Stein H, Hummel M. Histone
      ing in epidermotropic T-cell lymphomas. Am J Clin Pathol.                    acetylation and DNA demethylation of B cells result in a Hodgkin-
      2003;119(3):414–23.                                                          like phenotype. Leukemia. 2008;22:835–41.
                                 Mastocytosis
                                                                                                                                                      8
Mastocytosis comprises a group of clonal mast cell (MC)                Table 8.1 Updated WHO classification of mastocytosis 2016
disorders whose clinical course ranges from indolent to                 1. Cutaneous mastocytosis (CM):
aggressive. In the revised 2016 WHO classification, masto-               Urticaria pigmentosa (UP)/Maculopapular cutaneous mastocytosis
cytosis is included as a separate category and not any more                 (MPCM)
                                                                         Diffuse cutaneous mastocytosis
listed among myeloproliferative neoplasms [1, 2].
                                                                         Solitary mastocytoma of skin
    Non-neoplastic MCs are present in many human organs                 2. Indolent systemic mastocytosis (ISM)
including the lymphatic system and particularly abundant                 Meets criteria for systemic mastocytosis (SM). No “C” findings.
at major body interfaces with the external environment                      No evidence of associated hematological neoplasm
such as the skin, the lung, and the gastrointestinal tract                  Isolated bone marrow mastocytosis
[3]. MCs have pleiotropic functions including the multi-                 As above (ISM), but with bone marrow involvement and no skin
                                                                           involvement, generally low burden of MC
valent capacity to recognize and to react to internal and
                                                                        3. Smouldering systemic mastocytosis (SSM)
external dangers and form a bridge between innate and                    As above (ISM), but with 2 or more “B” findings, and no “C”
adaptive immunity. MCs store preformed mediators                           findings, 1 generally high burden of MC
within granules that are rapidly released thus promoting                4. Systemic mastocytosis with an associated hematological neoplasm
inflammation and local recruitment of other innate immu-                    (SM-AHN)
                                                                         Meets criteria for SM and criteria for AHN as a distinct entity per
nity cells [3].
                                                                            the WHO classification
    The spectrum of mastocytosis is heterogeneous and sub-              5. Aggressive systemic mastocytosis (ASM)
divided into cutaneous mastocytosis, systemic mastocytosis               Meets criteria for SM. One or more “C” findings. No evidence of
(SM), and localized mast cell tumors (Table 8.1). SM is                     mast cell leukemia
characterized by the accumulation of atypical MCs in one or             6. Mast cell leukemia (MCL)
more organ systems. The SM category includes indolent                    Meets criteria for SM. Bone marrow biopsy shows diffuse
                                                                            infiltration, usually dense, by atypical, immature mast cells. BM
SM, smouldering SM and SM with an associated hemato-                        aspirate smears show ≥20% mast cells. In classic cases, mast cells
logical neoplasm (SM-AHN), aggressive SM (ASM), and                         account for ≥10% of peripheral blood white cells. Aleukemic
mast cell leukemia (MCL) [4, 5]. The term advanced sys-                     MCL variant (<10% circulating mast cells)
temic mastocytosis is used for aggressive SM, SM-AHN,                   7. Mast cell sarcoma (MCS)
and mast cell leukemia. For the diagnosis of SM, one major               No evidence of SM. Generally localized destructive growth
                                                                            pattern. High-grade cytology
and several minor criteria have been defined. The major cri-
                                                                       Adopted from Horny et al. [2], Pardanani [5]
terion is the presence of multifocal clusters of abnormal              The diagnosis of SM can be made when the major criterion and one
≥15 MCs in the bone marrow or other organs. Minor diag-                minor criterion or at least three minor criteria are present (see Table 8.2)
nostic criteria include spindle-shaped or morphologically
Table 8.2 Major criterion and minor criteria, B and C findings that are      8.1     Epidemiology and Clinical Features
relevant for the classification of SM subtypes
Major criterion                                                              A prevalence of mastocytosis of 1 in 10,000 inhabitants has
 Multifocal, dense infiltrates of mast cells (≥15 mast cells in             been reported, but underdiagnosis is assumed [7]. CM pre-
  aggregates) detected in sections of bone marrow and/or other
  extracutaneous organ(s)
                                                                             dominantly affects children and may disappear during
Minor criteria                                                               puberty [8]. SM mastocytosis generally develops in adults
 (a) In biopsy sections of bone marrow or other extracutaneous             with a light male predominance [2].
       organs, >25% of the mast cells in the infiltrate are spindle-            The clinical features are variable and related to the sub-
       shaped or have atypical morphology or, of all mast cells in           type of mastocytosis. CM may present as urticaria pigmen-
       bone marrow aspirate smears, >25% are immature or atypical
 (b) Detection of an activating point mutation at codon 816 of KIT
                                                                             tosa, diffuse CM, or mastocytoma of skin.
       in bone marrow, blood, or other extracutaneous organ                     SM may be associated with four categories of
 (c) Mast cells in bone marrow, blood, or other extracutaneous             symptoms:
       organ express CD25 with/without CD2 in addition to normal
       mast cell markers. (Mast cell CD25 is the more sensitive
                                                                             (a) Constitutional symptoms (e.g., fatigue)
       marker, by both flow cytometry and immunohistochemistry)
 (d) Serum total tryptase persistently exceeds 20 ng/mL (unless            (b) Skin manifestations (e.g., urticarial)
       there is an associated myeloid neoplasm, in which case this           (c) Mediator-related symptoms (e.g., abdominal pain, gas-
       parameter is not valid)                                                   trointestinal problems)
“B” findings                                                                 (d) Musculo-skeletal symptoms (osteoporosis)
 1. High mast cell burden shown on BM biopsy: >30% infiltration
      of cellularity by mast cells (focal, dense aggregates) and serum
      total tryptase level > 200 ng/mL                                           Patients with advanced SM such as aggressive SM and
 2. Signs of dysplasia or myeloproliferation in non-mast cell              mast cell leukemia may develop hepato-splenomegaly and
      lineage(s), but insufficient criteria for definitive diagnosis of an   organ impairment for example liver failure secondary to
      associated hematological neoplasm (AHN), with normal or only           mast cell infiltrates. Bone marrow involvement may be asso-
      slightly abnormal blood counts.
                                                                             ciated with anemia, secondary eosinophilia, neutropenia,
 3. Hepatomegaly without impairment of liver function, palpable
      splenomegaly without hypersplenism, and/or lymphadenopathy             and thrombocytopenia or even bone marrow failure. In
      on palpation or imaging.                                               patients who develop SM-AHN, the outcome principally
“C” findings                                                                 depends on both disorders. However, the non-mast cell
 1. Bone marrow dysfunction caused by neoplastic mast cell                 hematologic neoplasm such as chronic myelomonocytic leu-
      infiltration, manifested by ≥1 cytopenia(s) (ANC <1.0 × 109/L,
     Hgb < 10 g/dL, and/or platelet count <100 × 109/L)
                                                                             kemia,      myelodysplastic/myeloproliferative     neoplasm,
 2. Palpable hepatomegaly with impairment of liver function,               unclassifiable, or acute myeloid leukemia often determines
      ascites, and/or portal hypertension                                    patient prognosis.
 3. Skeletal involvement with large osteolytic lesions with/without            Serum tryptase released from MCs is important for the
      pathological fractures (pathological fractures caused by               diagnosis and monitoring of SM. Total serum tryptase levels
      osteoporosis do not qualify as a “C” finding)
                                                                             ≥20 ng/mL are suggestive of SM. However, serum tryptase
 4. Palpable splenomegaly with hypersplenism
 5. Malabsorption with weight loss due to gastrointestinal mast cell       may be normal in patients with low MC burden, for example,
      infiltrates                                                            with indolent SM or isolated bone marrow mastocytosis [5].
Adopted from Horny et al. [2], Pardanani [5]                                 It is worth noting that tryptase levels may also be increased
                                                                             in non-MC neoplasms such as chronic myeloid leukemia.
They are predominantly localized in the perivascular and/or    mastocytosis by the revised 2016 WHO classification. In
paratrabecular bone marrow compartments and frequently         cases with minimal bone marrow involvement molecular
associated with focal reticulin fibrosis. MC aggregates may    analysis of laser microdissected MCS may be used to detect
be accompanied by reactive eosinophilia and/or lymphoid        the KIT D816V mutation. Mast cell leukemia may harbor
infiltrates. However, 20% of patients with indolent SM         atypical mutations in the KIT gene such as D816H/Y or
patients lack MC clusters in the bone marrow [5]. BM neo-      F522C [10]. Rare SM cases with normal morphologic and
plastic MCs are generally spindle-shaped with oval nuclei      immunophenotypic features may also harbor non-D816V
and elongated cytoplasmic processes. They may also have        KIT mutations such as germline F522C or somatic I817V
round nuclei and abundant clear cytoplasm. In high grade SM    mutations [5].
such as aggressive SM or MC leukemia, MCs may have bilo-          Additional mutations may be found especially in
bated or multilobated nuclei or even resemble to metachro-     advanced SM and include mutations in TET2, SRSF2,
matic blasts. MCs may easily be detected on Giemsa and/or      ASXL1, RUNX1, CBL, JAK2, and/or RAS genes [9]. There is
Naphthol-ASD-chloroacetate esterase stained bone marrow        evidence that mutations in SRSF2, ASXL1, RUNX1, and ele-
sections. However, since neoplastic MCs may contain rare       vated alkaline phosphatase are predictive adverse prognostic
metachromatic granules or may even be degranulated, they       markers for overall survival in SM [11]. Models for SM
may be missed on conventionally stained sections. Therefore,   based on clinical and integrated clinical-genetics informa-
immunohistochemical staining for tryptase and CD117 (c-kit)    tion have demonstrated the independent prognostic contri-
is mandatory when SM is suspected. However, these markers      bution of mutations [12].
do not discriminate between neoplastic and non-neoplastic
MCs. By contrast, the aberrant expression of CD25 by neo-
plastic MCs is considered as a hallmark of mastocytosis. CD2   8.4     Caution
immunostain is less helpful. Aberrant CD30 expression is
observed in subsets of neoplastic MCs especially in aggres-    • Mast cell infiltrates may easily be missed on H&E
sive SM and mast cell leukemia.                                  stained sections. The application of immunohistochemi-
    Mast cell leukemia is diagnosed when bone marrow aspi-       cal markers for assessment of mast cell infiltration is
rate smears contain ≥20% MCs that are generally immature,        mandatory.
atypical, and rather round than spindle-shaped. Mast cell      • Atypical mast cells especially in mast cell leukemia are
leukemia may be accompanied by a leukemic blood picture          frequently devoid of prominent polychromatic cytoplas-
with ≥10% peripheral blood MCs or be aleukemic.                  mic granules, may show lobulated nuclei, and may mimic
    A retrospective study of bone marrow biopsies based on a     monocytes/monoblasts.
cohort from the German Registry of Disorders on Eosinophils    • Expression of tryptase is not restricted to mast cells but
and Mast Cells has shown that SM may be overlooked by            may also be observed in basophilic leukemia that, how-
pathologists especially when immunohistochemical markers         ever, does not express CD117.
were not applied [6].                                          • Eosinophils may be abundant in mastocytosis so that
                                                                 myeloid/lymphoid neoplasms with eosinophilia and
                                                                 gene rearrangement may be considered as a differential
8.3      Genetics                                               diagnosis especially as these disorders often harbor a
                                                                 spindle-shaped CD25+ mast cell population. However,
Mutations in KIT, most frequently KIT D816V, are present in      mast cells in this category are generally loosely scattered
over 80% of all systemic mastocytosis patients [9]. The KIT      between eosinophils and rarely arranged in clusters ≥15
D816V mutation is listed as minor diagnostic criterion for       mast cells.
238                                                                                                                                   8   Mastocytosis
Case History 1. Bone Marrow Mastocytosis (Fig. 8.1)                             mast cell infiltrates accounting for about 15% of bone mar-
Routine laboratory examination of this 74-year-old man                          row cells. Aggregates were formed by >15 mast cells. The
revealed a mild chronic anemia. The CBC was as following:                       immunophenotype was characterized by a co-expression by
WBC 5.1 × 109/L, HB 11.3 g/dL, platelets 152 × 109/L. The                       CD25, CD117, and tryptase. CD2 was only expressed by
differential count was normal. No hepato-splenomegaly,                          rare lymphocytes. The mast cell infiltrates were associated
lymphodenopathy, or skin lesions were present. The serum                        with focal fibrosis. A focal moderate lacunar bone resorp-
tryptase level was 17 ng/mL. A bone marrow aspirate smear                       tion was observed but no large osteolytic lesions. A KIT
contained rare spindle-shaped mast cells containing abun-                       D816V mutation was present. The diagnosis was bone mar-
dant metachromatic granules. A bone marrow core biopsy                          row mastocytosis, a subtype of indolent systemic
was performed and showed paratrabecular and perivesicular                       mastocytosis.
a b
MGC Giemsa
c d
CD25 CD25
Fig. 8.1 Bone marrow mastocytosis. (a) Bone marrow aspirate smear               CD25; e, f, CD117). Note focal moderate lacunar bone resorption (b).
with spindle-shaped mast cells showing polychromatic cytoplasmic                (g, h) Perivascular and paratrabecular fibrosis associated with the mast
granulation (MGG). (b–f) Bone marrow core biopsy with paratrabecu-              cells (Gomori)
lar (b, e) and perivascular (c, d, f) mast cell infiltrates (b; Giemsa; c, d,
8       Mastocytosis                    239
e f
CD117 CD117
g h
Gomori Gomori
Case History 2. Aggressive Systemic Mastocytosis                          erate eosinophilia, and 9% spindle-shaped mast cells. The
(Fig. 8.2)                                                                bone marrow core biopsy showed focal replacement of
This 71-year-old male patient was admitted with hepato-                  hematopoiesis by sheets of MCs occupying about 35% of the
splenomegaly and pancytopenia. The CBC was as following:                  marrow. The serum tryptase level was 75 ng/mL. The serum
WBC 2.9 × 109/L, HB 9.3 g/dL, platelets 89 × 109/L. The dif-              alkaline phosphatase was 210 U/L. A KIT D816V mutation
ferential count was: neutrophils 41%, eosinophils 11%, mast               was present. The diagnosis was aggressive SM since 3 C
cells 1%, monocytes 2%, lymphocytes 45%. The bone mar-                    findings were present.
row aspirate showed reduced trilineage hematopoiesis, mod-
a b
MGG H&E
c d
H&E Gomori
Fig. 8.2 Aggressive systemic mastocytosis. (a) A rare round-shaped        fibrosis (d). Mast cells express CD25 (e) and trypase (f). (g) Naphthol-
mast cells with polychromatic cytoplasmic granules is detected in a       ASD-chloroacetate esterase+ mast cells are focally associated with
peripheral blood smear. (b, c) Bone marrow core biopsy showing large      lymphoid follicles. (h) Focal nearly complete replacement of myelopoi-
areas infiltrated by mast cells associated with eosinophilia (b, c) and   esis by MPO-negative mast cells (MPO)
8       Mastocytosis                      241
e f
                         CD25       Tryptase
    g                           h
NASDCL MPO
Case History 3. Systemic Mastocytosis with an                              with atypically lobulated nuclei and about 5% spindle-
Associated Hematologic Neoplasm (Myelodysplastic                           shaped mast cells. Myeloblasts arranged in small clusters
Syndrome with Excess Blasts 2 (SM-AHN, MDS-EB-2),                          represented 14% of nucleated bone marrow cells. Large
Fig. 8.3)                                                                   mast cell aggregates associated with fibrosis were detected
This 68-year-old male patient was admitted with marked                      in trephine biopsy and occupied about 20% of bone marrow
thrombocytopenia. The CBC was as follows: WBC                               area. They expressed CD25, CD117, and tryptase. The
11.8 × 109/L, HB 9.8 g/dL, MCV 86 fl, platelets                             serum tryptase level was 61 ng/mL. Mutational analysis
17 × 109/L. The differential count showed 54% neutrophils,                  revealed KIT D816V, SRSF2, and RUNX1 mutations. The
47% lymphocytes, and 9% myeloblasts. The bone marrow                        diagnosis was SM with an associated hematologic neoplasm,
aspirate smears were hypercellular and showed marked tri-                   myelodysplastic syndrome with excess blasts 2 (SM-AHN,
lineage dysplasia with numerous small megakaryocytes                        MDS-EB-2).
a b
MGG MGG
Fig. 8.3 Systemic mastocytosis with an associated hematologic neo-         (MGG). (d, e) Large sheets of spindle-shaped mast cells in the bone
plasm: myelodysplastic syndrome with excess blasts-2 (SM-AHN,              marrow trephine biopsy (H&E). Mast cells express CD117 (f) and
MDS-EB-2) (a–c) Bone marrow aspirate smears showing trilineage             trypase (g). Atypical localized clusters of myeloblasts are highlighted
hematopoietic dysplasia with numerous small hypolobulated mega-            by CD34 immunohistochemical stain (h)
karyocytes increased blast cells and scattered spindle-shaped mast cells
8       Mastocytosis                      243
c d
MGG H&E
e f
Giemsa CD117
g h
Tryptase CD34
Case History 4. Systemic Mastocytosis with an                               roblasts were observed. The bone marrow aspirate was mark-
Associated Hematologic Neoplasm: Acute Myeloid                              edly hypercellular and contained 65% CD34+ blasts that
Leukemia (SM-AHN, AML) (Fig. 8.4)                                           were partially positive for myeloperoxidase. Mast cells were
A 45-year-old male patient was admitted with marked pan-                    observed in bone marrow particles. Paratrabecular mast cell
cytopenia. The CBC was as follows: WBC 2.3 109/L, HB                        aggregates were detected in the bone marrow core biopsy. In
8.5 g/dL, platelets 8 × 109/L. The differential count showed                contrast to the myeloblasts, they were positive for Naphthol-
32% neutrophils, 3% metamyelocytes, 3% myeloblasts, 7%                      ASD-chloroacetate esterase, CD117, tryptase, and CD25.
monocytes, and 55% lymphocytes. In addition. some eryth-                       Genetic analysis revealed KIT D816V mutation.
a b
MGG MGG
c d
MPO Giemsa
Fig. 8.4 Systemic mastocytosis with an associated hematologic neo-          sheets of mast cells in the bone marrow core biopsy (Giemsa). (e–g) In
plasm: acute myeloid leukemia (SM-AHN, AML). (a, b) Bone aspirate           contrast to the AML blast population mast cells are positive for naph-
smears showing abundant small-sized mononuclear blasts. Note the            thol-ASD-chloroacetate esterase (e) and express CD117 (f) and trypt-
presence of spindle-shaped mast cells in bone marrow particles (MGG).       ase (g). (h) The AML blast population is immunolabeled by CD34
(c) Blasts are partially positive for myeloperoxidase. (d) Paratrabecular   antibody
8       Mastocytosis                     245
e f
NASDCL CD117
g h
Tryptase CD34
Case History 5. Systemic Mastocytosis with an                             by sheets of spindle-shaped CD25+ CD117+ tryptase+mast
Associated Hematologic Neoplasm: Acute Myeloid                            cells. They occupied about 15% of the marrow area and
Leukemia with Mutated NPM1 (SM-AHN, AML)                                  were associated with a slight increase in the reticulin
(Fig. 8.5)                                                                fibers. In contrast to the blasts, NPM1 immunostaining of
A 63-year-old woman presented with anemia and throm-                      mast cells was restricted to the nuclei. The serum tryptase
bocytopenia. The CBC was as follows: WBC 7.4 × 109/L,                     level was low. The karyotype obtained from cultured AML
HB 7.8 g/dL, MCV 86.3 fl, platelets 37 × 109/L. The dif-                  blasts was normal 46, XX. Genetic analysis by next-gener-
ferential count showed 37% neutrophils, 1% metamyelo-                     ation sequencing revealed DNMT3A, FLT3-ITD, and
cytes, 6% band forms, 8% blasts, 4% monocytes, and 44%                    NPM1 mutations but no KIT D816V mutation. This phe-
lymphocytes. The bone marrow aspirate contained 70%                       nomenon probably resulted from the low amount of mast
blasts that were positive for CD13, CD33, CD38, HLA-DR,                   cells. The diagnosis was systemic mastocytosis with an
and partially positive for MPO, CD11c, lysozyme, and                      associated hematologic neoplasm, AML with mutated
CD15. By cytologic examination, the blasts showed                         NPM1 (SM-AHN, AML).
myelomonocytic features. The bone marrow core biopsy                         The molecular analysis and the immunohistochemical
was diffusely infiltrated by myeloid blasts that showed an                NPM1 staining suggested no clonal relationship between the
atypical nuclear and cytoplasmic immunolabeling positiv-                  AML and the SM populations. It was discussed that the
ity by a NPM1 antibody as a surrogate marker of a NPM1                    AML but not the small SM component might be crucial for
mutation. In addition, paratrabecular areas were infiltrated              the prognosis of this patient.
a b
MGG MGG
Fig. 8.5 Systemic mastocytosis with an associated hematologic neo-        the bone marrow core biopsy adjacent to mononuclear blasts (Giemsa).
plasm: acute myeloid leukemia with mutated NPM1 (SM-AHN, AML).            (e, f) Mast cells express tryptase (e) and CD117 (f). (g, h) NPM1 immu-
(a) Presence of monoblasts and atypical myelomonocytic cells in the       nohistochemical stain shows a strictly nuclear positivity in mast cells
peripheral blood. (b) Abundant blasts and rare scattered mast cells are   (g) but a clearly aberrant cytoplasmic labeling of the AML population
present in bone marrow smears. (c) Blasts partially express myeloper-     (h) suggesting that the NPM1 mutation is restricted to AML blasts
oxidase. (d) Small sheets of spindle-shaped mast cells are detected in
8       Mastocytosis                      247
c d
MPO Giemsa
e f
Tryptase CD117
g h
NPM1 NPM1
Case History 10.6. Acute Mast Cell Leukemia (Figs. 8.6                 tially CD2, CD25, CD11c, and HLA-DR (30%), but negative
and 8.7)                                                               for CD3, CD7, CD14, CD15, CD19, CD34, CD38, CD56, and
A 44-year-old woman was admitted with leukocytosis, ane-               myeloperoxidase. Residual hematopoietic precursors showed
mia, and thrombocytopenia. The CPC was as follows: WBC                 dysplastic features. The bone marrow core biopsy was mark-
27.6 × 109/L, HB 8.3 g/dL, MCV 98 fl, platelets 49 × 109/L.            edly hypercellular and diffusely infiltrated by large blasts. The
   On peripheral blood smears, 28% large-sized blasts with             oval or slightly lobulated nuclei were rimed by a wide faintly
round or bilobated nuclei and sparse metachromatic cytoplas-           granulated eosinophilic cytoplasm. By immunohistochemical
mic granulation were detected. Some dysplastic neutrophils             stains, the atypical mast cells expressed CD117, tryptase,
and rare erythroblasts were also present. On bone marrow               CD25, and partially CD30. Serum tryptase level was 220 ng/
aspirate smears, large atypical mast cells represented 80% of          mL. Genetic analysis revealed a typical KITD816 mutation.
nucleated cells. The neoplastic population was positive for            The diagnosis was acute mast cell leukemia.
CD117 with co-expression of CD10, CD13, CD33, and par-
a b
MGG MGG
c d
MGG MGG
Fig. 8.6 Acute mast cell leukemia. (a–d) Peripheral blood smears       row aspirate smears with compact sheets and scattered highly atypical
showing large-sized blasts with round or bilobated nuclei and sparse   mast cells showing a slightly eosinophilic faintly granulated cytoplasm.
metachromatic cytoplasmic granulation. Note the presence of some       Residual erythropoiesis and myelopoiesis (f) and especially mega-
dysplastic neutrophils and rare erythroblasts MGG). (e–h) Bone mar-    karyocytopoiesis (g) with dysplastic features (MGG)
8       Mastocytosis                                                                                                                          249
e f
MGG MGG
g h
MGG MGG
a b
H&E H&E
Fig. 8.7 Acute mast cell leukemia. (a, b) Extensive infiltration of the   esterase (c) and myeloperoxidase (d) while both reactions stained resid-
bone marrow core biopsy by atypical mast cells showing oval or slightly   ual myeloid cells. (e–h) The atypical mast cells expressed CD117 (e),
lobulated nuclei and a wide faintly granulated eosinophilic cytoplasm     tryptase (f), partially CD30 (g) and CD25 (h)
(H&E). (c, d) Mast cells were negative for naphthol-ASD-chloroacetate-
250                                 8   Mastocytosis
c d
NASDCL MPO
e f
CD117 Tryptase
g h
CD30 CD25
9.1       Introduction                                                adolescents are diagnosed with this disease. The male to
                                                                       female ratio is 1.4:1–2.2:1.
MPNs comprise various groups of clonal hematopoietic stem                 Since the late years of the twentieth century, treatment regi-
cell disorders. The WHO classification 2017 subdivides MPNs            mens have been revolutionized. With the detection of imatinib
into BCR-ABL1-positive chronic myeloid leukaemia (CML)                 and the development of second- and third-generation of TK
and BCR-ABL1-negative or Philadelphia (Ph) chromosome                  inhibitors, the 5-year survival of CML patients is more than 90%.
negative MPNs. BCR-ABL1-negative MPNs are PV, ET,
PMF, CNL and CEL-NOS as well as MPN unclassifiable.
    Since the WHO classification 2008 (fourth edition), the            9.2.2      Morphology
categories in the 2017 revision of the WHO classification of
myeloid neoplasms did not undergo significant changes [1].             Usually a bone marrow biopsy is performed to evaluate the
The effort of the updated revised WHO classification is to             phase of CML and to exclude other diseases with leukocyto-
incorporate new prognostic, clinical, morphologic, immuno-             sis. Furthermore, histology of the bone marrow is the only
phenotypic, and genetic data that have been detected since             option to establish a bone marrow fibrosis.
the last edition. Newly identified molecular features can
influence diagnosis and prognosis of MPNs, especially
BCR-ABL1-MPN. Furthermore, the reproducibility of the                 9.2.3      Chronic Phase (CP)
diagnoses has been improved due to the standardization of
various variable morphologic features.                                 In most cases the bone marrow is highly hypercellular due to
    In PV patients, JAK2 mutations were found which were in            a highly increased granulocytopoiesis. The paratrabecular
more than 95% JAK2 V617F mutations and in the remaining                and perivascular cuff of immature granulopoietic cells is
patients exon 12 mutations. In ET patients, JAK2 V617F,                thickened with up to 10 cells, while in a normal bone mar-
CALR, and MPL mutations were observed in 53.4%, 21.9%,                 row, this layer comprises 2–3 cells. Centripetally, the granu-
and 1.7% of patients, respectively; the remaining ET cases             locytopoiesis is maturing with masses of segmented
were triple negative. In PMF patients, JAK2 V617F, CALR,               granulocytes in the center of the marrow spaces. An increase
and MPL mutations were present in 67.5%, 13.7%, and 2.6%               of eosinophils and basophils is quite common. The blast
of patients, respectively; the remaining cases were triple             count in the chronic phase is ≤5%.
negative [2].                                                              The megakaryocytes in most cases are increased; however,
                                                                       in some cases the number of megakaryocytes can be normal
                                                                       or even decreased. Megakaryocytes in CML are smaller than
9.2        hronic Myeloid Leukemia (CML),
          C                                                            normal megakaryocytes and show hypolobulated nuclei.
          BCR-ABL1-Positive                                            These megakaryocytes are called “dwarf” forms.
                                                                           Erythropoiesis is decreased in most cases. The erythrones
9.2.1     Epidemiology                                                are smaller than normal and also the erythroid cells are smaller.
                                                                       Two thirds of patients with CML lack fibrosis in the first
The annual incidence of CML in Europe ranges from 1 to 2               biopsy, however up to 30% of CML-patients are first diag-
cases per 100,000 inhabitants [3]. CML is mainly a disease             nosed with a moderate to marked reticulin fibrosis (grade 2 or
of adults (median age is about 50 years), rarely children and          3), and a few patients even show secondary bone formation.
   Often, abnormal macrophages, like sea-blue histiocytes       Table 9.1 Criteria for CML, accelerated phase (according to the 2016
and pseudo-Gaucher cells can be found in the marrow. These      revision to the World Health Organization classification of myeloid
                                                                neoplasms and acute leukemia [12])
cells result from an increased cell turnover and are derived
from the neoplastic clone [4].                                  CML, accelerated phase criteria
                                                                Any one or more of the following hematologic/cytogenetic criteria or
   Since the tyrosine kinase inhibitor (TKI) therapy newly      response-to-TKI criteria:
diagnosed CML patients have a nearly normal life span [5],      • Persistent or increasing WBC            “Provisional” response-to-
but regular monitoring for BCR-ABL1 burden is essential            (>10 × 109/L), unresponsive to          TKI criteria
to find disease progression and development to TKI resis-          therapy
tance. The pathologist must be aware of the particular treat-   • Persistent or increasing                • Hematologic resistance
                                                                   splenomegaly, unresponsive to              to the first TKI (or
ment the patient has received when asked to evaluate the           therapy                                    failure to achieve a
response to the therapy. Even though monitoring of the                                                        complete hematologic
therapy response is mainly done by chromosome and                                                             responsea to the first
molecular studies, the pathologic findings in a bone mar-                                                     TKI) or
                                                                • Persistent thrombocytosis               • Any hematological,
row biopsy contribute to the accurate assessment of the            (>1000 × 109/L), unresponsive to           cytogenetic, or molecular
patient’s status [1].                                              therapy                                    indications of resistance
                                                                                                              to two sequential TKIs or
9.2.3.1 Morphologic Findings with TKI Therapy                  • Persistent thrombocytopenia             • Occurrence of two or
After 3 months, patients responsive to TKI therapy show            (<100 × 109/L) unrelated to therapy        more mutations in
                                                                                                              BCR-ABL1 during TKI
nearly normal bone marrows. There is a complete hemato-                                                       therapy
logic response with the reduction of bone marrow cellularity    • 20% or more basophils in the PB
and correction of the M:E ratio, and normalization of mega-     • 10–19% blastsb in the PB and/or BM
karyocyte size. After treatment, reactive lymphoid aggre-       • Additional clonal chromosomal
gates consisting of mainly small B- and T-lymphocytes are          abnormalities in Ph+ cells at diagnosis
                                                                   that include “major route”
frequently found. If, during therapy, changes in the hemato-       abnormalities (second Ph, trisomy 8,
logic or morphologic features are obvious (like elevation of       isochromosome 17q, trisomy 19),
WBC count, re-appearance of basophilia and dwarf mega-             complex karyotype, or abnormalities
karyocytes, development of marrow fibrosis), this most             of 3q26.2
                                                                • Any new clonal chromosomal
likely indicates a loss or failure of response, demanding          abnormality in Ph+ cells that occurs
cytogenetic and/or molecular investigations to determine the       during therapy
causes.                                                         Large clusters or sheets of small, abnormal megakaryocytes, associated
                                                                with marked reticulin or collagen fibrosis in biopsy specimens may be
9.2.3.2 Disease Progression of CML                             considered as presumptive evidence of AP, although these findings are
                                                                usually associated with one or more of the criteria listed above
The natural history of CML (without treatment) is transfor-     a
                                                                 Complete hematologic response: WBC, <10 × 109/L; platelet count,
mation to a blastic transformation of any one or multiple       <450 × 109/L, no immature granulocytes in the differential count and
hematopoietic cell lines.                                       spleen non-palpable
                                                                b
                                                                  The finding of bona fide lymphoblasts in the blood or marrow, even if
                                                                <10%, should prompt concern that lymphoblastic transformation may
                                                                be imminent and warrants further clinical and genetic investigation;
9.2.4   Accelerated Phase (AP)                                 20% or more blasts in blood or BM, or an infiltrative proliferation of
                                                                blasts in an extramedullary site is CML, blast phase
Disease progression to AP has been reduced to 1–1.5% per
year from more than 20% per year in the pre-TKI era [6].
The criteria of the AP vary in the literature. In the revised   9.2.5     Blast Crisis (BC)
WHO classification, criteria for AP include morphologic,
hematologic and cytogenetic parameters, additional impacts      The diagnosis of a BC is defined by at least 20% blasts in the
of genetic evolution, and evidence to resistance to TKIs (see   peripheral blood or the bone marrow or an extramedullary
Table 9.1).                                                     infiltration of blasts. In more than two thirds of patients, a
   Histologically, in bone marrow biopsy, the most conspic-     myeloid BC is evident, which is usually accompanied or pre-
uous features are the increase of basophils and the increase    ceded by markedly increased basophils. Patients can develop
of blasts. Basophils can be detected reliably by metachro-      a lymphoid BC showing an immunophenotype identical to
matic stained sections (e.g., MGG) and/or immunohisto-          ALL. The revised WHO classification 2017 has a new defini-
chemically (e.g., antibody 2D7). Blasts are most likely         tion for lymphoid blast crisis, stating that any lymphoblasts in
detected immunohistochemically (e.g., CD34).                    the PB should raise concern for BC and cases with >5% lym-
9   Myeloproliferative Neoplasm (MPN)                                                                                       255
Case History 1 (CML CP) (Fig. 9.1)                                          The histology of the bone marrow biopsy showed highly
A 54-year-old female presented with abdominal pain and                   hypercellular bone marrow with highly increased granulocy-
pain at the spinal column. The splenic size was increased with           topoiesis. Paratrabecular and perivascular broad sheets of
a diameter of 15 cm. Further medical work-up revealed leu-               immature myeloid precursors were evident. There was a
kocytosis with 129.83 × 109/L, anemia with Ery 3.00 × 1012/L,            marked increase of small megakaryocytes diffusely distrib-
Hb 8.6 g/dL, and thrombocytosis with 565 × 109/L.                        uted. Erythropoietic cells were decreased.
   Peripheral blood, bone marrow cytology, and bone mar-
row histology as well as cytogenetic evaluation revealed
BCR/ABL-positive CML, CP.
a b
c d
CD15 MPO
Fig. 9.1 Case 1 (CML, CP). (a) Highly hypercellular bone marrow is       spaces. (e, f) Marked increase of small megakaryocytes (so-called
seen in routinely stained H&E section. (b) Thickened perivascular cuff   dwarf forms) stained with an antibody to CD42b. (g) Nucleated eryth-
of immature granulopoietic cells (H&E). (c, d) Immunohistochemistry      ropoietic cells are decreased (glycophorin C). (h) Using an antibody to
using myeloid markers (CD15, MPO) show a marked increase of gran-        CD34 no increase of blasts is found
ulocytopoiesis with maturation toward the central areas of the marrow
9   Myeloproliferative Neoplasm (MPN)                            257
e f
CD42b CD42b
g h
Glycophorin C EryCD34
Case History 2 (CML, CP) (Fig. 9.2)                                        Further work-up of peripheral blood, bone marrow, and
During a routine control, a 64-year-old male patient was found          cytogenetics revealed BCR-ABL1-positive CML, CP with
to have a pathological blood count (Ery 3.68 T/L, leukocytes            fibrosis.
15.14 × 109/L, thrombocytes 193 × 109/L). The patient was oth-
erwise symptom-free; the spleen was only slightly enlarged.
a b
c d
GOMORI GOMORI
Fig. 9.2 Case 2 (CML CP). (a, b) Routinely stained H&E sections         Immunohistochemically, no increase of basophils is seen (2D7). (g)
show a moderate to highly hypercellular bone marrow with up to 20%      Only a slight increase of granulopoietic cells can be detected (MPO).
fat cells. (c, d) The marrow spaces show a variable fibrosis which is   (h) Characteristically, an increase of small megakaryocytes (CD42b) is
graded according to WHO classification as grade 2 (e, f)                evident
9   Myeloproliferative Neoplasm (MPN)                259
e f
2D7 2D7
g h
MPO CD42b
Case History 3 (CML AP) (Fig. 9.3)                                         positive blasts was evident (blast count was 15%).
A 65-year-old female with treated CML for 16 years pre-                    Furthermore, immunohistochemically an increase of
sented with fever and pathological blood picture as fol-                   basophils could be detected. The diagnosis was “highly
lows: leukocytes: 23.61 G/L, Ery 4.08 T/L, Hb 12.4 g/dL,                   hypercellular bone marrow with diffuse reticulin- and col-
Hkt 35.8% and Thrombo 46 G/L. Bone marrow biopsy                           lagen fibrosis (grade 2) and increase of basophils and
was highly hypercellular with diffuse reticulin fibrosis                   blasts according to accelerated phase of the known BCR-
grade 2 and collagen fibrosis grade 2. Small megakaryo-                    ABL1-positive CML.
cytes were markedly increased, and an increase of CD34-
a b
c d
GOMORI GOMORI
Fig. 9.3 Case 3 (CML, AP). (a, b) Routinely stained H&E sections           Increase of basophils highlighted with an antibody to 2D7. (f) Mature
show highly hypercellular bone marrow with marked increase of small        granulocytes, stained with an antibody to CD15 are decreased. (g)
megakaryocytes. (c, d) Staining for fibers (Gomori’s stain) reveals dif-   Increase of CD34-positive blasts with an amount of 15%. (h) Masses of
fuse reticulin as well as multifocal collagen fibrosis (grade 2). (e)      small megakaryocytes (CD42b) are present
9       Myeloproliferative Neoplasm (MPN)                 261
e f
                                             2D7       CD15
    g                                              h
CD34 CD42b
Case History 4 (CML, BC-AML) (Figs. 9.4 and 9.5)                               33.2 g/dL, and Thrombo 20 G/L. A bone marrow biopsy
A 40-year-old male with treated CML for 8 years presented                      showed irregular cellularity with marked fibrosis and
with pancytopenia: leukocytes: 3.41 G/L, Ery 3.22 T/L, Hb                      increase of more than 30% myeloid blasts, thus myeloid
12.9 g/dL, Hkt 38.8%, MCV 120.5 fL, MCH 40.1 pg, MCHC                          blast crisis was diagnosed.
a b
c d
Fig. 9.4 Case 4 (CML, BC-AML). (a) Variable cellularity of the bone            of small megakaryocytes. (e–h) The highly hypercellular, as well as the
marrow spaces (H&E). (b–d) In the highly hypercellular marrow                  hypocellular marrow spaces reveal a dense fibrosis (reticulin: grade 3,
spaces, a diffuse infiltration of blasts is seen, in addition to an increase   collagen: grade 3) as shown with Gomori’s stain
9   Myeloproliferative Neoplasm (MPN)                                                                                                           263
e f
GOM GOM
g h
GOM GOM
a b
MPO CD33
Fig. 9.5 Case 4 (CML, BC-AML). (a) Most blasts are reactive with an         ity of blasts are reactive with an antibody to CD34. (f) Strong specific
antibody to MPO. (b) Using an antibody to CD33, most blasts show            CD7-positivity of the myeloid blasts. (g, h) High number of small
specific reactivity. (c) Most blasts are CD15-positive. (d, e) The major-   megakaryocytes (dwarf forms) stained with an antibody to CD61
264                               9   Myeloproliferative Neoplasm (MPN)
c d
CD15 CD34
e f
CD34 CD7
g h
CD61 CD61
Case History 5 (CML, BC-ALL) (Fig. 9.6)                                  with Leuko 6.89 G/L, Ery 1.84 T/L, Hb 5.8 g/dL, Hkt 16.3%,
A 60-year-old female with Alzheimer’s disease and treated                Thrombo 43 G/L, and LDH 340 U/L. The bone marrow
BCR-ABL1+ CML for 2 years was admitted to the hospital                  biopsy showed lymphoid BC.
because of a fainting fit. The blood picture was pathologic
a b
c d
CD10 CD20
Fig. 9.6 Case 5 (CML, BC-ALL). Lymphatic BC CML. H&E stained             for CD10 (c), CD20 (d), CD34 (e) and TdT (f). (g, h) Morphologically,
section shows a highly hypercellular bone marrow (a) with dense infil-   and also by immunohistochemistry, no CD42b-positive megakaryo-
tration of blast (b) with round nuclei and several nucleoli (inset).     cytes and no MPO-positive granulopoietic cells could be identified
Immunohistochemistry revealed infiltration by lymphoid blasts positive
266                                9   Myeloproliferative Neoplasm (MPN)
e f
CD34 TdT
g h
CD42b MPO
a b
                                                            H&E                                                                  H&E
c                                                                     d
EPX CD61
Fig. 9.7 Unusual morphology of CML. Highly hypercellular bone         with masses of small megakaryocytes (e: H&E; f: CD42b). (g) Biopsy
marrow biopsy of a 76-year-old woman with BCR-ABL1-positive           of a 42-year-old male patient with BCR-ABL1-positive CML with
CML with marked eosinophilia (a, b: H&E). Eosinophilia is verified    many fat cells (MGG). (h) Pretreatment bone marrow biopsy of a
with an antibody to EPX (c). Small megakaryocytes, typical for BCR-   28-year-old woman with BCR-ABL1-positive CML with marked fibro-
ABL1-positive CML are stained with an antibody to CD61 (d). Bone     sis and secondary bone formation (MGG)
marrow biopsy of a 64-year-old female with BCR-ABL1-positive CML
268                                                                                      9   Myeloproliferative Neoplasm (MPN)
e f
                                                        H&E                                                          CD42b
g                                                                h
MGG MGG
• Occasionally, in pretreatment bone marrow biopsies of          • Occasionally, the differential diagnosis includes
  patients with BCR-ABL1+ CML, a variable amount of                CNL, a rare disorder with neutrophilia, splenomegaly,
  fatty tissue can be seen (Fig. 9.7g shows the biopsy of a        and hepatomegaly. CNL is characterized by CSF3R
  42-year-old male patient).                                       mutation, and in peripheral blood and the bone mar-
• In rare instances, pretreatment BCR-ABL1-positive CML            row, mature granulocytic cells without a left shift are
  presents with marked fibrosis and osteosclerosis as is shown     seen.
  in Fig. 9.7h. This pretreatment bone marrow originates from    • Cave, there are several confusing terms:
  a 28-year-old woman. Despite intensive treatment with TKI        –– A “Ph-negative CML”: this is CML without Ph chro-
  and bone marrow transplantation, the woman developed                mosome but BCR/ABL1+. Before the area of molecu-
  blast crisis and died 2.5 years after primary diagnosis.            lar technologies, this included as well BCR/ABL1+ as
• CML is the most common MPN associated with thrombo-                 BCR/ABL1− cases.
  cytosis. When the thrombocytosis is quite extreme, ET            –– B “atypical CML”: this is NOT CML (there is no Ph
  might be simulated.                                                 chromosome, and there is no BCR/ABL1).
• Rare cases of p190 BCR-ABL + CML have monocytosis                   Morphologically, prominent granulocytic dysplasia
  and may mimic CMML. In these patients, a second rear-               and often multilineage dysplasia are obvious. Usually,
  rangement, minor breakpoint cluster region (m-bcr),                 few basophils are seen in atypical CML.
  occurs between the first intron of the bcr gene and exon 2       –– C “juvenile CML”: this is an old terminus, but this is
  of the abl gene, producing the e1a2 transcript that codes           NOT CML (there is no Ph chromosome, no BCR/
  for a 190 kDa protein. This fusion transcript occurs in             ABL1). The disease is now called “JCMML” (juvenile
  1–2% of CML patients as a sole rearrangement [9].                   CMML).
9   Myeloproliferative Neoplasm (MPN)                                                                                                  269
a b
c d
Fig. 9.8 CNL. (a, b) The bone marrow is highly hypercellular for the        section, normal erythroid islands are seen, occasionally disrupted by
age with a cellularity of 95%. The sinusoids are expanded (MGG). (c)        the granulopoietic cell proliferation. Megakaryocytes are normal in
The maturation of the highly increased granulopoiesis is normal with        number and do not show atypia. (e) Highly increased granulopoiesis
immature myeloid cells located paratrabecular and perivascular. There       stained with an antibody to CD15. (f) Erythropoiesis, stained with an
is no relative increase in myeloblasts, mast cells, or eosinophils (MGG).   antibody to glycophorin A, does not show dysplastic features
(d) No reticulin fibers are found (Gomori’s stain). In Gomori’s stained
9   Myeloproliferative Neoplasm (MPN)                                                                                   271
e f
CD15 Glycoph.A
9.3.5    Caution                                              kemoid reaction of the bone marrow may show a similar
                                                               histology.
The clonality of the neutrophilic cell population has to be        CNL is a rare, but predictable, complication of SAA. This
established cytogenetically or by molecular techniques; oth-   is related to the proliferative pressure of SAA on hematopoi-
erwise, correctly making the diagnosis of CNL can be chal-     etic stem cells. The integrative diagnosis is supported by
lenging for pathologists and clinicians alike.                 NGS.
    Without knowledge of genetic abnormalities, the diagno-        Allogenic stem cell transplantation is the only curative
sis of CNL can be suggested, but not confirmed, since a leu-   option.
272                                                                                                9   Myeloproliferative Neoplasm (MPN)
a b
c d
MGG
Fig. 9.9 PV (masked). (a–c) H&E stained bone marrow biopsy shows            increase in reticulin fibers is visible (Gomori’s stain). (f) No iron depos-
highly hypercellular bone marrow with increase of all hematopoietic         its can be found (BBL). (g) Immunohistochemically stained erythropoi-
cells. Often, as in this case, in PV dilated vessels can be seen. (d) The   esis is markedly increased (CD71). (h) Increased granulopoiesis is
increased megakaryocytes are highly polymorphous (MGG). (e) No              stained immunohistochemically with an antibody to MPO
274                                                                                      9   Myeloproliferative Neoplasm (MPN)
e f
GOM BBL
g h
CD71 MPO
9.4.4     Caution                                                    Some patients have “masked PV.” This term was re-
                                                                   introduced for JAK2-mutated patients with latent (ini-
The presence of reticulin fibrosis does not exclude the diag-      tial, occult pre-polycythemic) disease manifestations,
nosis of PV.                                                       but who present with a BM morphology consistent with
   If iron deposits are found, the patient might have a bleeding   PV [18].
history. Otherwise, the diagnosis of PV has to be reassessed.
9   Myeloproliferative Neoplasm (MPN)                                                                                                    275
9.5      Primary Myelofibrosis (PMF)                            correct diagnosis of prefibrotic PMF is the atypical mega-
                                                                 karyocytopoiesis. There are clusters of megakaryocytes,
9.5.1    Epidemiology                                           often located paratrabecular or perisinusoidal, and most
                                                                 megakaryocytes are enlarged, but there are also small forms.
The incidence rate of PMF ranges from 0.1 per 100,000 per        The nuclei often show clumped chromatin, and the nuclei
year to 1 per 100,000 per year [19]. The median age at diag-     appear “cloud-like” with hypolobulation. Naked megakaryo-
nosis is 65 years with a preponderance for males.                cytic nuclei are often seen. Lymphoid nodules are not uncom-
                                                                 mon and usually the bone marrow shows vascular
                                                                 proliferation. Most patients with prefibrotic PMF gradually
                                                                 transform into overt PMF. The diagnostic criteria for diagno-
9.5.2    Morphology                                             sis of pre PMF are listed in the following table.
In addition to morphology of the bone marrow biopsy, driver          WHO prePMF criteria [1, 15]
                                                                     Major criteria
mutations are integrated into WHO diagnostic criteria. The
                                                                     1. Megakaryocytic proliferation and atypia, without reticulin fibrosis
mutational frequency in PMF is 58% JAK2, 25% CALR, and                      >grade 1, accompanied by increased age-adjusted BM cellularity,
7% MPL [15].                                                                granulocytic proliferation, and often decreased erythropoiesis
                                                                      2. Not meeting the WHO criteria for BCR-ABL1+ CML, PV, ET,
                                                                            myelodysplastic syndromes, or other myeloid neoplasms
                                                                       3. Presence of JAK2, CALR, or MPL mutation or in the absence of
9.5.3    PMF, Prefibrotic/Early Stage                                      these mutations, presence of another clonal marker, or absence of
                                                                            minor reactive BM reticulin fibrosis
About 30–40% of patients with PMF are diagnosed in the           Minor criteria
early stage without showing a significant reticulin or colla-    Presence of at least one of the following, confirmed in two
gen fibrosis (grade 0 or 1). The bone marrow biopsy is           consecutive determinations:
                                                                 a. Anemia not attributed to a comorbid condition
hypercellular with hyperplastic granulopoiesis and increase
                                                                  b. Leukocytosis ≥11 × 109/L
 of atypical megakaryocytes. There may be a slight left shift      c. Palpable splenomegaly
 of granulopoietic cells without an increase of myeloblasts in      d. LDH increased to above upper normal limit of institutional
 percentage. There is no increase of CD34+ blasts. Usually,                 reference range
 erythropoiesis is decreased, but some patients show rather          Diagnosis of prePMF requires meeting all three major criteria and at
 prominent erythroid precursor cells. The key feature for a          least one minor criterion
276                                                                                                       9   Myeloproliferative Neoplasm (MPN)
Case History 1 (Pre-PMF)                                                    24.2/pg, MCHC 33.3 g/dL, Thrombo 810 G/l, LDH
A 74-year-old female was admitted to the hospital because                   288 U/l.
of cardiac insufficiency. During the medical work-up, a                        These findings were clinically suspicious for PV, addi-
pathologic blood picture, slightly increased LDH and                        tionally JAK2-positivity was found. A bone marrow
slight splenomegaly were found: Leuko 12.0 G/l, Ery                         biopsy was performed and diagnosed as MPN, pre-PMF
5.99 T/l, Hb 14.5 g/dL, Hkt 43.6%, MCV 72.8/L fL, MCH                       (Fig. 9.10).
a b
c d
GOM
Fig. 9.10 (a) The bone marrow is highly hypercellular with only a few       (d) or show a slight circumscribed fibrosis (e). (f) Immunohistochemically
irregularly distributed fat cells (H&E). The most striking feature is the   stained megakaryocytes (CD42b) are markedly increased with cluster
increased, highly polymorphous and atypical megakaryocytopoiesis            formation. (g) Large atypical megakaryocytes with hypolobulated
with cluster formation (b) and hypolobulated nuclei with dense chro-        nuclei forming a cluster (CD61). (h) The marrow reveals rather promi-
matin (b, c). Most marrow spaces are either devoid of reticulin fibers      nent erythroid precursor cells (CD71)
9       Myeloproliferative Neoplasm (MPN)                 277
e f
GOM CD42b
g h
CD61 CD71
9.5.4    PMF, Overt Fibrotic Stage                                        3. Presence of JAK2, CALR, or MPL mutation or in the absence of
                                                                                 these mutations, presence of another clonal marker, or absence of
                                                                                 reactive myelofibrosis
In this stage the bone marrow shows marked reticulin fibro-
                                                                            Minor criteria
sis (grades 2 and 3) and frequently also collagen fibrosis of               Presence of at least one of the following, confirmed in two
various extents. Cellularity is quite variable. In one and the              consecutive determinations:
same biopsy, there can be areas with hypercellularity and                   a. Anemia not attributed to a comorbid condition
marked hypocellularity. In most cases the bone marrow is                     b. Leukocytosis ≥11 × 109/L
hypocellular for age with foci of hematopoietic cells alter-            c. Palpable splenomegaly
                                                                         d. LDH increased to above upper normal limit of institutional
nating with fatty tissue or fibers. Megakaryocytes are highly
                                                                                 reference range
atypical and can be arranged in dense sheets. Often mega-                 e. Leukoerythroblastosis
karyocytes are present within dilated sinusoids. Some                      Diagnosis of overt PMF requires meeting all three major criteria and
patients show dense fibrosis and nearly no hematopoietic                   at least one minor criterion
cells or a few immature hematopoietic islands within ves-
sels. Secondary bone formation of various degrees can be
detected [1, 20].
    Disease acceleration is diagnosed, if patients with previ-          9.5.5     Immunohistochemistry
ously assured PMF show 10–19% blasts in the peripheral blood
or bone marrow, or if there is an increase of CD34+ blasts              There are no documented abnormal phenotypic features.
detected immunohistochemically. Acceleration of overt PMF               However, in PMF (early stage and fibrotic stage) immuno-
may additionally be indicated by an increase of monocytoid              histochemistry is helpful for detecting the atypical mega-
differentiated cells [21]. Transformation to acute leukaemia is         karyocytes, which are highlighted by using markers for
indicated by the finding of ≥20% blasts. The diagnostic criteria        megakaryocytes, such as CD41, CD42b, or CD61.
for PMF, fibrotic stage are listed in the following table.              Furthermore, the exact number of erythropoietic cells can
                                                                        be estimated with antibodies to CD71, glycophorin A or
                                                                        gylycophorin C. Also the amount of granulopoietic cells
WHO overt PMF criteria [1, 15]                                          and an increase of blasts might need immunohistochemical
Major criteria
                                                                        investigation (CD15, MPO, CD33, CD34, CD117c).
1. Presence of megakaryocytic proliferation and atypia, accompanied
      by either reticulin and/or collagen fibrosis grades 2 or 3        Recently, an antibody to calreticulin has been developed.
 2. Not meeting WHO criteria for ET, PV, BCR-ABL1+ CML,               The use of this antibody allows a specific and rapid detec-
      myelodysplastic syndromes, or other myeloid neoplasms             tion of calreticulin mutations [22].
9       Myeloproliferative Neoplasm (MPN)                                                                                                        279
Case History (PMF Overt Stage)                                             Hb 10.3 g/dL, Hkt 31.3%, MCV 92.3 fL, MCH 30.4 pg,
A 52-year-old male was admitted to the hospital because of                 MCHC 32.9 g/dL, Thrombo 98 G/L. LDH was increased
back pain, anemia, and thrombocytopenia. Furthermore the                   with LDH 486 U/l. JAK2 was detected.
patient complained about night sweats since 6 weeks without                   A bone marrow biopsy was performed revealing MPN,
infectious disease. The spleen was markedly enlarged. The                  overt PMF (reticulin fibrosis grade 2, collagen fibrosis grade
blood count was as following: Leuko 7.73 G/L, Ery 3.39 T/L,                2 and osteosclerosis grade 2) (Fig. 9.11).
a b
c d
GOM
Fig. 9.11 Overt PMF. (a–c) In H&E stained sections, the bone marrow        lin fibrosis grade 2 and focal collagen fibrosis (gold-colored fibers vis-
is markedly hypercellular. The bone trabeculae focally are irregularly     ible in (e). (f) Immunohistochemically, with an antibody to CD61, there
formed with newly formed bone (osteosclerosis grade 2). This second-       is a marked increase of atypical megakaryocytes with focal cluster for-
ary bone formation is seen as focal budding, hooks, spikes, and focal      mation. (g) Erythropoietic cells are decreased and irregularly distrib-
paratrabecular apposition of new bone. Striking increase of atypical       uted (CD71). (h) Granulopoietic cells (stained immunohistochemically
megakaryocytes is evident. (d, e) Gomori’s stain reveals diffuse reticu-   with MPO) are still prominent
280                                                                                     9   Myeloproliferative Neoplasm (MPN)
e f
GOM CD61
g h
CD71 MPO
Case History (ET)                                                         The bone marrow biopsy was normocellular with normal
A 54-year-old male was admitted to the hospital after an               appearing erythro- and granulocytopoiesis. There was a mod-
accident with rib fracture. During work-up, marked throm-              erate increase of normal sized and enlarged megakaryocytes
bocytosis (937 G/L) was found. All remaining blood test                with often markedly hyperlobulated nuclei. Occasionally,
results were normal, and the patient had no history of throm-          loose clusters of megakaryocytes were found. No fibrosis was
bosis or other complaints. JAK2 mutation was found and a               seen. The diagnosis was MPN, ET (Fig. 9.12).
bone marrow biopsy was performed.
a b
c d
Fig. 9.12 ET (a) Normocellular bone marrow. (b) No pathological        seen (MGG). (f–h) Rather large megakaryocytes with hyperlobulated
fibrosis is seen in Gomori’s stain. (c) Megakaryocytes are increased   (“stag-horn” like) nuclei (MGG)
(MGG). (d, e) Loose cluster of pleomorphic megakaryocytes can be
9       Myeloproliferative Neoplasm (MPN)       283
e f
g h
a b
c d
Fig. 9.13 Addendum to MPN: PV, PMF, and ET. Since megakaryocytes             like nuclei as well as cluster-formation (e, f: H&E; g, h: MGG). (i–l)
are the hallmark cells in MPNs, figures of megakaryocytes in the bone mar-   Megakaryocytes in ET: Most megakaryocytes are enlarged with abundant
row of these diseases are shown: Megakaryocytes in PV are highly pleo-       cytoplasm and hyperlobulated, occasionally “stag-horn”-like (l) nuclei.
morphic and are of different size. Marked maturation defects concerning      These megakaryocytes are often arranged in loose clusters (i, j: MGG).
the nuclear-cytoplasmic differentiation are not seen (a, b: H&E; c, d:       Megakaryocytes in post-PV-MF (m, n, H&E) and post-ET-MF (o, p,
MGG). Megakaryocytes in PMF are highly atypical with abnormal nuclear-       H&E) are highly pleomorphic and atypical with hyper- or hypolobulated
cytoplasmic ratio, abnormal clumping of chromatin and occasional cloud-      nuclei and often clumped chromatin
9       Myeloproliferative Neoplasm (MPN)       285
e f
g h
i j
k l
m n
o p
a b
c d
GOM BBL
Fig. 9.14 (a) Representative bone marrow biopsy with highly hyper-             topoiesis can be seen (MPO). (f) Erythropoiesis (CD71) is slightly to
cellular bone marrow (MGG). (b) Increase of all hematopoietic cells            moderately increased with a few enlarged (megaloblastic appearing)
can be seen even in H&E stained section. (c) No increase of reticulin          cells. The megakaryocytes are increased and polymorphic; some mega-
fibers is evident. (d) Iron stain (Berlin Blue) detects a few iron granules.   karyocytes show hypolobulated nuclei (g: MGG, h: CD61)
(e) Immunohistochemically a marked increase of maturing granulocy-
9   Myeloproliferative Neoplasm (MPN)                                                                          289
e f
MPO CD71
g h
CD61
a b
CD61
Fig. 9.15 Monocytoid progression of PV. (a) The first biopsy shows         Erythrocytopoiesis is decreased and dysplastic. A few small atypical
highly hypercellular bone marrow with the characteristic morphology        megakaryocytes are seen in this marrow space (H&E). (e, f) Increase of
of PV with increase of all hematopoietic cell lines (H&E). (b) First       monocytoid cells stained with an antibody to CD14. (g) Slight increase
biopsy with increased polymorphic megakaryocytes (CD61). (c)               (>5%) of CD34-positive blasts. (h) Left side: In some areas large mega-
Second biopsy 2 years later: There is highly increased cellularity with-   karyocytes, typical for PV can be seen, other areas (right side) show
out fat cells (H&E). (d) Second biopsy: Higher magnification with          quite small atypical megakaryocytes which usually are seen in cases of
highly increased granulocytopoiesis with increased monocytoid forms.       CMML (MGG)
9       Myeloproliferative Neoplasm (MPN)                291
c d
e f
CD14 CD14
g h
CD34
a b
MGG MGG
c d
MGG MPO
Fig. 9.16 CEL-NOS. Highly hypercellular bone marrow with marked         appearing erythropoiesis (CD71). (f) Megakaryocytes are not increased,
eosinophilia with maturation (a, b, c: MGG). (d) Granulopoietic cells   they are rather small but not atypical (CD61)
immunohistochemically stained with an antibody to MPO. (e) Normal
294                                                                                     9   Myeloproliferative Neoplasm (MPN)
e f
CD71 CD61
9.9.4    Caution                                              have to suggest CEL-NOS, but the diagnosis is only possible
                                                               with a complete knowledge of clinical and genetic results.
If the bone marrow is hypercellular with marked eosinophilia      In cases with eosinophilia also consider reactive eosino-
and normal appearing erythro- and megakaryocytopoiesis, you    philia (e.g., allergic) and idiopathic HES.
9   Myeloproliferative Neoplasm (MPN)                                                                                                      295
Myeloid/lymphoid neoplasms (M/LN) with eosinophilia and                Table 10.1 Myeloid/lymphoid neoplasms with eosinophilia and rear-
gene rearrangement are listed in a distinct WHO category.              rangement of PDGFRA, PDGFRB, or FGFR1, or with PCM1-JAK2a
This category includes three specific disease groups involv-               Diagnostic criteria of a MPN, AML, or ALL/LBL with eosinophilia
                                                                           associated with FIP1L1-PDGFRA
ing rearrangement of platelet-derived growth factor receptor
                                                                           A myeloid or lymphoid neoplasm, usually with prominent
α (PDGFRA), platelet-derived growth factor receptor β                      eosinophilia and a FIP1L1-PDGFRA fusion gene resulting from a
(PDGFRB), fibroblast growth factor receptor 1 (FGFR1) or                   cryptic del(4)(q12) or a variant fusion gene with rearrangement of
is associated with PCM1-JAK2, a provisional WHO entity                     PDGFRA or clinic-pathologic surrogate markers
[1–3] (Table 10.1). The presentation of the disorders is het-              Diagnostic criteria for myeloid/lymphoid neoplasms associated with
                                                                           ETV6-PDGFRB fusion gene or other rearrangement of PDGFRB
erogeneous: Phenotypic features may vary between those of
                                                                           A M/LN myeloid or lymphoid neoplasm, often with prominent
a myeloproliferative neoplasm (MPN), especially chronic                    eosinophilia and sometimes with neutrophilia or monocytosis and
eosinophilic leukemia (CEL), myelodysplastic syndrome/                     presence of t(5;12)(q32;p13.2) or a variant translocation or
MPN (MDS/MPN), or occasionally of an acute myeloid leu-                    demonstration of an ETV6-PDGFRB fusion gene or rearrangement
kemia (AML), a myeloid sarcoma (MS), or an acute lympho-                   of PDGFRB. Molecular confirmation is highly desirable. Cases with
                                                                           fusion genes typically associated only with BCR-ABL1-like
blastic leukemia/lymphoma (ALL/LBL). It has been                           B-lineage ALL are excluded
strengthened that although the classification of this category             Diagnostic criteria of MPN or acute leukemia associated with
of neoplasms is predominantly based on genetic aberrations,                FGFR1 rearrangement (also referred to as 8p11myeloproliferative
diagnosis should still be anchored to a combination of histo-              syndrome)
                                                                           A MPN or MDS/MPN neoplasm with prominent eosinophilia, and
morphology and clinical and laboratory criteria [3].
                                                                           sometimes with neutrophilia or monocytosis or acute myeloid
                                                                           leukemia or precursor T-ALL/LBL or precursor B-ALL/LBL or
                                                                           mixed phenotype acute leukemia (usually associated with peripheral
10.1      Epidemiology and Clinical Features                              blood or BM eosinophilia) and presence of t(8;13)(p11.2;q12.1) or a
                                                                           variant translocation leading to FGFR1 rearrangement demonstrated
                                                                           in myeloid cells, lymphoblasts, or both
M/LN with eosinophilia and gene rearrangement represent                    Diagnostic criteria for myeloid/lymphoid neoplasms with
rare disorders. A striking male predominance is reported in                PCM1-JAK2
neoplasms with PDGFRA, PDGFRB rearrangements, and                          A myeloid or lymphoid neoplasm, often with prominent eosinophilia
with PCM1-JAK2 while the M/LN with FGFR1 rearrange-                        and presence of t(8;9)(p22;p24.1) or a variant translocation leading
                                                                           to JAK2 rearrangement
ment has a male-to-female ratio of 1.5:1 [2]. The age range
                                                                       a
                                                                        Adapted and modified from [3]
of the patients is wide ranging from the first to eighth decade
in all types.
    Patients with M/LN with PDGFRA rearrangement gener-
ally present as a chronic MPN with eosinophilia. Peripheral               M/LN associated with ETV6-PDGFRB fusion gene or
blood and bone marrow eosinophilia are frequently associ-              other rearrangement of PDGFRB include several variants.
ated eosinophilia-related secondary effects such as pruritus,          Patients often present with eosinophilia, spleno ± hepato-
splenomegaly, and/or organ fibrosis. Serum tryptase is often           megaly, occasionally skin involvement and/or cardiac fail-
mildly elevated (>12 ng/mL). The disease is responsive to              ure. Serum tryptase may be mildly or moderately elevated.
tyrosine kinase inhibition, especially imatinib. Rare patients         However, eosinophilia is not always present.
develop AML or T-ALL/LBL [4].
patients, the two most frequently observed cytogenetic          MPN such as atypical BCR-ABL1-negative chronic myeloid
abnormalities were t(8;13)(p11.2;q12)(partner gene              leukemia.
ZMYM2) and t(8;22)(p11.2; q11.2)(BCR). The large major-
ity of patients had a RUNX1 mutation, and all developed an
acute leukemia [9].                                             10.4     Caution
   PCM1-JAK2 as a consequence of t(8;9)(p22;p24.1) is
the most frequent JAK2 fusion gene in this extremely rare       • Blood and bone marrow eosinophilia may be inconspicu-
provisional WHO entity [3, 10]. Since both myeloid and            ous or even absent in some cases harboring the typical
lymphoid lineages may be involved in PCM1-JAK2-positive           mutational spectrum of the category M/LN with eosino-
neoplasms, it has been suggested that the disorder origi-         philia and gene rearrangement.
nates from an early pluripotent hematopoietic progenitor or     • Patients may initially present with T-/ or rarely B-ALL/
stem cell [3]. Only a few patients with t(9p24;v)/JAK2 at         LBL mimicking precursor lymphoid neoplasms NOS.
diagnosis have been reported. In a common study from            • The presence of atypical CD25+ spindle-shaped mast
large medical centers, most patients with 9p24/JAK2 pre-          cells and the increased serum tryptase levels may hint to
sented with MPN-associated features characterized by vari-        systemic mastocytosis associated with eosinophilia.
able degrees of eosinophilia, myelofibrosis, frequent           • M/LN with eosinophilia and rearrangement of PDGFRA
proliferations of early erythroblasts in bone marrow and          are extremely rare in female patients. Therefore, this
extramedullary sites, and infrequent/absent somatic muta-         diagnosis assigned to a woman should be further
tions [10]. They may also present with features of a MDS/         discussed.
300                10 Myeloid/Lymphoid Neoplasms with Eosinophilia and Rearrangement of PDGFRA, PDGFRB, FGFR1, or with PCM1-JAK2
Case History 1. Myeloid/Lymphoid Neoplasm with                            showed a marked proliferation of left-shifted eosinophils and
Eosinophilia and Rearrangement of PDGFRA (Fig. 10.1)                      rare neutrophil precursors. Spindle-shaped mast cells coex-
A 57-year-old male patient consulted a dermatologist with                 pressing tryptase, CD117, and CD25 were scattered among
pruritus and fatigue. The CBC was as follows: WBC                         eosinophils. No mast cell aggregates were present.
25 × 109/L, Hb 7.3 g/dL, RBC 1.9 × 1012/L, platelets                      Immunohistochemical stain showed nuclear positivity for
98 × 109/L. The peripheral blood smears showed 58% pre-                   phospho(p)-SAT5. Reverse transcriptase polymerase chain
dominantly mature but partially degranulated eosinophils, 9%              reaction (RT-PCR) revealed a FIP1L1-PDGFRA fusion gene.
neutrophils, 3% myelocytes, 4% band forms, and 17% lym-                   The diagnosis was myeloid/lymphoid neoplasm with
phocytes. The bone marrow core biopsy was hypercellular and               PDGFRA rearrangement.
a b
Giemsa Giemsa
c d
NASDCL MPO
Fig. 10.1 Myeloid/lymphoid neoplasm with eosinophilia and rear-           scattered through the marrow spaces. (f) Slight increase in reticulin
rangement of PDGFRA. (a, b) The bone marrow core biopsy is strik-         (Gomori stain). (g) Presence of atypical spindle-shaped mast cells is
ingly hypercellular and contains about 80% eosinophils of all stages of   highlighted by tryptase immunolabeling. (h) Nuclear expression pat-
maturation (Giemsa). (c) Naphthol-ASD-chloroacetate-esterase reac-        tern of pSTAT5. The diagnosis was myeloid/lymphoid neoplasm with
tion and (d) myeloperoxidase immunolabeling performed on the FFPE         eosinophilia and rearrangement of PDGFRA presenting with features
marrow sections. (e) Erythropoiesis is not arranged in a typical island   of chronic eosinophilic leukemia
10 Myeloid/Lymphoid Neoplasms with Eosinophilia and Rearrangement of PDGFRA, PDGFRB, FGFR1, or with PCM1-JAK2       301
e f
HB Gomori
g h
Tryptase pSTAT5
Case History 2. Myeloid/Lymphoid Neoplasm with                             were present. Monocytes were increased. Atypically local-
Eosinophilia and Rearrangement of PDGFRA (Fig. 10.2)                       ized aggregates of CD34+ blasts accounting for about 6%
A 49-year-old male was admitted with fatigue, weight loss,                 of nucleated cells were observed. Megakaryocytes were
and bone pain. The CBC was as follows: WBC 19 × 109/L,                     rarely observed and dysplastic. Immunostain for pSTAT5
Hb 9.1 g/dL, MCV 111 fL, platelets 98 × 109/L. The periph-                 showed a marked nuclear positivity of myeloid cells. In
eral blood smears showed 45% eosinophils, 16% dysplastic                   addition, scattered spindle-shaped mast cells with an aber-
hypolobulated neutrophils, 1% blasts, 1% myelocytes, 2%                    rant tryptase+ CD117+ CD25+ phenotype were detected.
band forms, 14% monocytes, and 18% lymphocytes. The                        The massive erythroid hyperplasia was suspicious for
bone marrow aspirate resulted in a dry tap. The markedly                   PCM1-JAK2. However, molecular testing by RT-PCR pro-
hypercellular bone marrow core biopsy showed a dense                       vided evidence of PDGFRA rearrangement. The diagnosis
reticulin fiber network consistent with fibrosis grade 2.                  was myeloid/lymphoid neoplasm with eosinophilia and
Erythropoietic cells formed large sheets. Dysplastic eosin-                rearrangement of PDGFRA mimicking chronic myelo-
ophils and neutrophil granulocytes and their precursors                    monocytic leukemia.
a b
PAS Gomori
c d
HB CD34
Fig. 10.2 Myeloid/lymphoid neoplasm with eosinophilia and rear-            blasts. (e) Biebrich scarlet reaction further provides evidence of signifi-
rangement of PDGFRA. (a) Aspect of the hypercellular bone marrow           cant eosinophilia. (f) CD68+ macrophages as well as monocytes are
core biopsy at low magnification (PAS). (b) Dense reticulin fiber net-     abundant. (g) Strong nuclear expression of pSTAT 5 by myeloid cells.
work consistent with fibrosis grade 2 (Gomori stain). (c) Large atypical   (h) Scattered spindle shaped tryptase+ mast cells
sheets of hemoglobin+ erythoid precursors. (d) Aggregates of CD34+
10 Myeloid/Lymphoid Neoplasms with Eosinophilia and Rearrangement of PDGFRA, PDGFRB, FGFR1, or with PCM1-JAK2        303
e f
g h
pSTAT5 Tryptase
Case History 3. Myeloid/Lymphoid Neoplasm with                              expressing CD25 and CD2 were observed. No BCR-ABL, KIT-
Eosinophilia and Rearrangement of PDGFRA                                    D816V, JAKV617F, calreticulin, or MPL mutation were present
Simultaneously Presenting as Chronic Eosinophilic                           while screening FIP1L1-PDGFRA rearrangement was positive.
Leukemia and T-Lymphoblastic Lymphoma (Figs. 10.3                               In addition, a lymph node biopsy was performed and
and 10.4)                                                                   showed a completely different microscopic aspect:
This 48-year-old male patient consulted his physician because                   The lymph node was diffusely infiltrated by small-sized
he had observed a rapidly growing cervical lymph node. The                  blasts exhibiting a grey-blue cytoplasm. The blast cell popula-
CBC was as follows: WBC 32 × 109/L, Hb 8.7 g/dL, MCV                        tion expressed cytoplasmic CD3, CD1a, CD4, CD8, CD99,
97 fL, platelets 82 × 109/L. The peripheral blood smears showed             TdT and showed a high Ki67+ proliferation fraction >90%.
38% eosinophils, 24% neutrophils, 6% metamyelocytes, 5%                     Eosinophils were not increased, but scattered spindle-shaped
myelocytes, 2% monocytes, and 25% lymphocytes. Because of                   atypical mast cells were noted. The immunophenotype sug-
a dry tap, no bone marrow aspirate smear was available. The                 gested a cortical T-lymphoblastic lymphoma. Clonal T-cell
hypercellular bone marrow core biopsy showed a marked prolif-               receptor γ rearrangement was detected by PCR analysis. Similar
eration of eosinophils at all stages of maturation, a left-shifted          to the bone marrow testing a FIP1L1-PDGFRA rearrangement
increased granulopoiesis and about 6% 34+ blast cells. Erythroid            was detected by FISH. The diagnosis was myeloid/lymphoid
colonies were atypically localized and diminished. Rare pre-                neoplasm with eosinophilia and rearrangement of PDGFRA
dominantly small megakaryocytes were present. In addition, a                presenting as CEL in bone marrow and peripheral blood and as
low number of spindle-shaped tryptase+ CD117+ mast cells co-                T-lymphoblastic lymphoma in the lymph node.
a b
                                                             Giemsa                                                                     Giemsa
                                                                                                                                        G
                                                                                                                                        Giie
                                                                                                                                           em
                                                                                                                                            msa
                                                                                                                                             sa
c d
Giemsa MPO
Fig. 10.3 Myeloid/lymphoid neoplasm with eosinophilia and rear-             myeloperoxidase. (e, f) Scattered spindle-shaped mast cells positive for
rangement of PDGFRA simultaneously presenting as chronic eosino-            CD117 (e) and for CD25 (f). (g) Atypical localized CD71+ erythroid
philic leukemia and T-lymphoblastic lymphoma: Bone marrow core              colonies. (h) PDGFRA rearrangement demonstrated by FISH using a
biopsy. (a–c) Hypercellular bone marrow core biopsy showing a               break apart probe. Note the presence of one separated green and one
marked proliferation of eosinophils at all stages of maturation (Giemsa).   separate orange signal in addition to a green/orange fusion signal
(d) Left-shifted increased myelopoiesis highlighted by immunostain for
10 Myeloid/Lymphoid Neoplasms with Eosinophilia and Rearrangement of PDGFRA, PDGFRB, FGFR1, or with PCM1-JAK2                               305
e f
CD117 CD25
g h
                                                                 CD71
                                                                 CD
                                                                 CD71
                                                                   71                                                              PDGFRA
a b
Giemsa CD1a
Fig. 10.4 Myeloid/lymphoid neoplasm with eosinophilia and rear-             plasmic CD3 (c), TdT (d), and CD99 (e). (f) Presence of scattered
rangement of PDGFRA simultaneously presenting as chronic eosino-            spindle-shaped CD117+ mast cells. (g, h) PDGFRA rearrangement
philic leukemia and T-lymphoblastic lymphoma: Lymph node biopsy.            demonstrated by FISH using a break apart probe. Note the presence of
(a) Diffuse infiltration of a lymph node by small-sized blasts exhibiting   one separated green and one separate orange signal in addition to a
a grey-blue cytoplasm. Strong positivity of blasts for CD1a (b) cyto-       green/orange fusion signal
306               10 Myeloid/Lymphoid Neoplasms with Eosinophilia and Rearrangement of PDGFRA, PDGFRB, FGFR1, or with PCM1-JAK2
c d
CD3 TdT
e f
CD99 CD117
g h
PDGFRA PDGFRA
Case History 4. Myeloid/Lymphoid Neoplasm with                           CD34 labeled endothelial cells but did not provide evidence
Eosinophilia and Rearrangement of FGFR1 (Fig. 10.5)                      of increased blasts. Numerous spindle-shaped atypical mast
This 39-year-old male patient complained about pruritus and              cells were highlighted by tryptase, CD117, and CD25 immu-
fatigue. Clinical examination revealed splenomegaly. The                 nolabeling. They were loosely arranged but did not form
CBC was as following: WBC 27 × 109/L, Hb 9.3 g/dL, MCV                   dense clusters. Moreover, nodular lymphoid infiltrates com-
89 fL, platelets 94 × 109/L. The peripheral blood smears                 posed of small inconspicuous B- and T-cells were present.
showed 35% partially hypogranulated eosinophils, 28% neu-                No clonal T-cell receptor γ or β and immunoglobulin heavy
trophils, 3% myelocytes, 2% band forms, 9% monocytes,                    chain rearrangements were present. FISH analysis for
and 23% lymphocytes. Eosinophilia was also a prominent                   PDGFRA and PDGFRB rearrangements were negative.
feature observed in the bone marrow core biopsy. Neutrophil              However, FGFR1 rearrangement was detected by further
precursors were also increased while erythropoiesis was                  screening by FISH. The diagnosis was myeloid/lymphoid
diminished. Megakaryocytes showed dysplastic features.                   neoplasm with eosinophilia and rearrangement of FGFR1.
a b
c d
MGG CD20
Fig. 10.5 Myeloid/lymphoid neoplasm with eosinophilia and rear-          Lymphoid nodules composed of CD20+ B-cells (d) and of CD3+
rangement of FGFR1. (a) Hypercellular bone marrow core biopsy            T-cells (e). (f) Evidence of numerous loosely arranged CD117+ spindle-
nearly devoid of fat cells showing two nodular lymphoid infiltrates      shaped atypical mast cells. (g) CD34 decorates predominantly vascular
adjacent to trabecular bone (H&E). (b) Increased neutrophil precursors   endothelial cells. No excess of blasts. (h) CD15 expression by the left-
and numerous dysplastic megakaryocytes (H&E). (c) Markedly               shifted myeloid lineage
increased eosinophils at different stages of maturation (MGG).
308               10 Myeloid/Lymphoid Neoplasms with Eosinophilia and Rearrangement of PDGFRA, PDGFRB, FGFR1, or with PCM1-JAK2
e f
CD3 CD117
g h
CD34 CD15
Clinical symptoms include fatigue, night sweats, weight            Cytogenetic aberrations involving many different chromo-
loss, and hepatosplenomegaly, especially in cases with             somes have been reported in 30–40% patients. Examples are
higher monocyte counts. Extramedullary disease involving           trisomy 8, trisomy 10, del(5q), monosomy 7 and complex
the skin for example may be present [3, 4].                        karyotypes [2, 3].
                                                                       Molecular alterations are more frequent and detected in
                                                                   about 90% of patients. The molecular profile is complex and
11.2.3 Bone Marrow and Blood Findings                             includes mutations in genes involved in methylation (TET2,
                                                                   50–60%), RNA splicing (SFSR2, 36–46%; U2AF35,
The BM is generally hypercellular with a focally accentuated       5–15%), histone modification (ASXL1, 43–44%; EZH2,
expansion of myelomonocytic cells. They show slightly              6–10%), transcription (RUNX1, 15–20%; CEBPA, 4–20%),
folded nuclei and a rather broad pale cytoplasm.                   signaling pathways (CBL, 10–20%; KRAS, 7–11%; NRAS,
Promonocytes are characterized by a fine stippled chromatin        4–16%; SETBP1, 6–16%; JAK2, 10%) [2].
structure and distinct nucleoli and abundant light gray or
slightly basophilic cytoplasm. These typical features may
better be detected in BM smears than in sections [6].              11.2.6 Prognosis
Dysplasia may involve one or more myeloid lineages.
Micromegakaryocytes are often the hallmark of dysmega-          Several prognostic scoring systems have been proposed
karyopoiesis. Reticulin fibrosis is reported in about one third to evaluate the overall survival with special consideration
of the cases and associated with a shorter median time to       for the risk of transformation into acute myeloid leuke-
disease progression [8–10]. Since fibrosis grade was shown      mia. These scores consider different variables such as
to be an important indicator of prognosis, with high-grade      age, monocytosis, circulating blasts, anemia, thrombocy-
fibrosis predicting inferior survival, it has been proposed thattopenia, fibrosis, and genetic findings [8, 10]. A moderate
fibrosis grading should be incorporated into prognostic         to severe myelofibrosis was reported in a 3.1% of CMML
assessment and therapeutic decision-making [9].                 cohort and associated with worse overall survival [9, 10].
                                                                Fifty percent of these patients had JAK2 p.V617F muta-
                                                                tion [10].
11.2.4 Immunohistochemistry Applied to BM                         It is well accepted that somatic mutations are involved in
         Trephine Biopsies                                      the heterogeneous prognosis of CMML 10 [11]. In a large
                                                                CMML cohort, mutations in ASXL1, RUNX1, NRAS, and
Myelomonocytic cells of CMML are positive for CD33, SETBP1 were important molecular prognostic factors. In a
myeloperoxidase (MPO), often for naphthol-ASD-multivariable regression model, a CMML-specific
chloroacetate esterase and show variable expressions of Prognostic Scoring System-Mol was defined which inte-
CD14, CD68, CD163, overexpression of CD56, and aberrant grated the genetic score with RBC transfusion requirement,
expression of CD2 and CD65. Small nodules of plasmacy- WBC and BM blast counts, and was able to identify four
toid dendritic cells within the marrow are highlighted by risk groups with significantly different survival and risk of
CD123 staining and are considered as a part of the malignant leukemic evolution [12]. In a cohort of CMML patients
clone.                                                          treated with hypomethylating agents TET2mut/ASXL1wt gen-
   Special point: An increase in CD34+ may be a hallmark otype predicted higher complete response rate and pro-
of disease progression. However, the evaluation of CD34+ longed survival while RUNX1mut and CBLmut genotypes were
BM cells is no reliable tool to detect the percentage of blasts associated with poorer outcome, independently of higher
and promonocytes since they often do not express CD34.          leukocyte count [11].
11   Myelodysplastic/Myeloproliferative Neoplasms (MDS/MPN)                                                          313
Case History 1 (CMML-1) (Figs. 11.1 and 11.2)                            decreased but showed dysplastic features including micro-
A 66-year-old male patient presented with reduced physical               forms. The erythropoiesis was quantitatively decreased
performance state, fatigue, and enlarged spleen size (Fig. 11.1).        with mild dyserythropoiesis. The granulopoiesis was left
   Laboratory findings: Hb 9.5 g/dL, platelets 110 × 109/L,              shifted. CD34-positive precursor/blast cells and promono-
WBC 25 × 109/L. The differential count showed 38% mono-                  cytes represented about 8% of nucleated bone marrow
cytes, 1% promonocytes, 45% neutrophils, 1% eosinophils,                 cells.
and 15% lymphocytes.                                                        The karyotype was normal. Next-generation sequencing
   The BM smears and the trephine BM biopsy were                         revealed a TET2 mutation. The diagnosis was CMML-1 pre-
hypercellular with rare fat cells. The myelomonocytic lin-               senting with a proliferative phenotype.
eage expressing myeloperoxidase and CD33 accounted for                      Case 1, Follow-up (CMML, Splenectomy)
more than 90% of all nucleated cells. Monocytes express-                    Because of abdominal discomfort and increasing symp-
ing CD14 and partially CD68 and CD56 were increased to                   tomatic thrombocytopenia splenectomy was performed
about 20%. Promonocytes and blast cells accounted for                    showing a dense infiltration of the red pulp by myelomono-
about 8% of BM cells. The megakaryopoiesis was not                       cytic cells (Fig. 11.2).
a b
                                                                 HE                                                                        HE
  c                                                                       d
CD14 CD34
Fig. 11.1 Bone marrow core biopsy (a–e), peripheral blood smear (f),     of small hypolobated CD61-positive dysplastic megakaryocytes. (f)
and bone marrow aspirate smears (g, h) from the 66-year-old patient      Peripheral blood smear showing mature monocytes and monocyte pre-
diagnosed with chronic myelomonocytic leukemia-1 (CMML-1). (a, b)        cursors as well as dysplastic granulocytes and (g) bone marrow aspirate
Hypercellular marrow spaces showing proliferation of myeloid and         characterized by a proliferation of myeloid and monocytic lineages and
monocytic cells (H&E). (c) The monocytic lineage is immunolabeled        rare erythroid cells (MGG). (h) Alpha-naphthyl acetate esterase stain-
by CD14 antibody. (d) Rare CD34-positive precursor cells. (e) Presence   ing highlighting the monocytic lineage
11    Myelodysplastic/Myeloproliferative Neoplasms (MDS/MPN)                                                                       315
e f
CD61 MGG
g h
MGG ANAE
a b
HE HE
Fig. 11.2 Splenectomy specimen from the patient with chronic myelomonocytic leukemia (CMML) showing predominant infiltration of the red
pulp by myelomonocytic cells (a–c H&E) that are partially positive for NASDCL (d, e), CD33 (f), and CD14 (g, h)
316                                   11   Myelodysplastic/Myeloproliferative Neoplasms (MDS/MPN)
c d
HE NASDCL
e f
NASDCL CD33
g h
                          CD
                          CD1
                            14
                          CD144                                                           CD1
                                                                                          CD
                                                                                          CD14
                                                                                             14
                                                                                              4
Case History 2 (CMML-2) (Figs. 11.3 and 11.4)                            monocytic lineages and 8% were eosinophils. Erythropoiesis
A 63-year-old lady was admitted with anemia (Hb 8.2 g/dL),               and megakaryopoiesis were significantly decreased. By CD61
leukocytosis (WBC 52 × 109/L), and thrombocytopenia                      immunohistochemistry, micromegakaryocytes and rare mega-
(platelets 67 × 109/L). The differential count showed aniso-             karyoblasts were detected. Promonocytes, monoblasts, and
cytosis of red blood cells, 32% monocytes, 7% promono-                   CD34-positive blasts accounted for about 17% of nucleated
cytes, 4% myelocytes, 3% metamyelocytes, 14% band                        bone marrow cells consistent with CMML-2.
forms, 39% neutrophils, 1% blasts, 8% eosinophils, and 13%                  Case 2, Follow-up (CMML, Progression to AML)
lymphocytes.                                                             (Fig. 11.4).
   Immunophenotyping of peripheral blood revealed neutro-                   Seven months later, the condition of the patient deterio-
phils with an aberrant expression pattern of CD13-CD16 and               rated. Promonocytes, monoblasts, and myeloblasts repre-
CD11b-CD13, monocytes with a reduced expression of                       sented 60% of peripheral blood and 85% of bone marrow
CD14, CD45, HLA-DR, and a coexpression of CD56.                          cells consistent with a progression into an acute myeloid leu-
Molecular analysis by next-generation sequencing reported                kemia (AML) that was classified as AML with myelodyspla-
negativity for BCR-ABL but mutations in ASXL1, EZH2, and                 sia related changes according to 2016 WHO and fulfilled
TET2 genes.                                                              morphologic criteria of acute myelomonocytic leukemia
   Sections of the hypercellular BM core biopsy (Fig. 11.3)              according to the previous French-American British classifi-
showed that 85% of nucleated cells belonged to the myeloid and           cation [7].
a b
                                                               H&E
                                                               H&
                                                                &E                                                                   H&E
 c                                                                       d
                                                           Giiem
                                                           G  emssa
                                                                  a                                                            NASDCL
                                                                                                                               NA
                                                                                                                               NASSD
                                                                                                                                   DCL
                                                                                                                                    C
Fig. 11.3 Chronic myelomonocytic leukemia-2 (CMML-2).                    NASDCL). (e) Strikingly diminished megakaryopoiesis with rare
Proliferation of the myeloid and monocytic lineage with an increase in   CD61-positive micromegakaryocytes and megakaryoblasts. (f)
immature precursors including promonocytes and monoblasts associ-        Scattered CD71-positive predominantly immature erythroid cells. (g,
ated with an expansion of eosinophils (a, b H&E, c Giemsa, d             h) Focal increase of CD34-positive myeloblasts
318                                                                         11   Myelodysplastic/Myeloproliferative Neoplasms (MDS/MPN)
e f
CD61 CD71
g h
                                                             CD34
                                                             C
                                                             CD
                                                              D334
                                                                 4                                                                       CD34
                                                                                                                                         CD
                                                                                                                                         CD3
                                                                                                                                           34
                                                                                                                                            4
a b
MGG ANAE
Fig. 11.4 Transformation of CMML-2 in overt acute myeloid leuke-        is densely infiltrated by basophilic blast cells (d, e) that are partially
mia (AML). Peripheral blood smears show myelomonocytic blast cells      NASDCL-positive (f) and frequently express CD34 (g, h). Note that a
with (a) slightly indented immature nuclei and a basophilic cytoplasm   small dysplastic megakaryocyte is also labeled by the CD34 antibody
(MGG) staining for ANAE (b) and partially MPO (c). The core biopsy      (h)
11   Myelodysplastic/Myeloproliferative Neoplasms (MDS/MPN)                319
c d
                                                        MP
                                                        M PO          Giiems
                                                                      G  ems
                                                                         em sa
                                                                             a
e f
                                                     Giie
                                                     G  emm
                                                          msa
                                                            sa
                                                            sa       NA
                                                                     N AS
                                                                        SDDC
                                                                           CLL
 g                                                               h
                                                       CD
                                                       C D
                                                         D334
                                                           34           CD
                                                                        C D34
                                                                           34
Case History 3 (Myelodysplastic Syndrome Evolving                     gene (ex3 and ex7), and in the JAK2 gene (ex14
into CMML, JAK2 V617F Mutated with Fibrosis,                          c.1849G > T p.V617F). A mixture of abundant small dys-
Further Progressing to AML) (Figs. 11.5, 11.6, and 11.7)              plastic and large polymorphic megakaryocytes reminis-
Initially, a hematologic work-up of this 56-year-old male             cent of a myeloproliferative neoplasm was a hallmark of a
patient was done because of thrombocytopenia. CBC: WBC                newly performed bone marrow biopsy. Abundant imma-
4.2 × 109/L, hemoglobin 10.2 g/dL, MCV 88 fL, platelet                ture myeloid and monocytic precursors with an increase in
count 45 × 109/L. The peripheral blood smear showed mild              promonocytes and monoblasts to 12% and dysplastic ery-
granulocytic dysplasia and monocytosis (16%). The bone                throid islands were observed. Reticulin fibrosis grade 1
marrow aspirate and core biopsy were hypercellular with a             had developed (Fig. 11.6). The diagnosis was chronic
proliferation of myeloid and monocytic precursors and more            myelomonocytic leukemia-2 with fibrosis grade 1 associ-
mature forms and mild erythroid dysplasia. Megakaryocytes             ated with a JAK2 pV16F mutation and myeloproliferative
were clearly dysplastic with a predominance of small dys-             features.
plastic forms. No fibrosis was observed (Fig. 11.5). The                 Case 3, Transformation into AML (Fig. 11.7):
karyotype was normal. No BCR-ABL1 or JAK2 pV16F muta-                    The patient’s clinical condition and hematologic parame-
tion was found. The diagnosis was myelodysplastic syndrome            ters rapidly deteriorated and acute myeloid leukemia devel-
with multilineage dysplasia, evolution to chronic myelo-              oped. Three months later, the CPC was: WBC 95 × 109/L
monocytic leukemia possible. The patient refused treatment.           (86% blasts) hemoglobin 8.4 g/dL, platelet count
   Case 3, Follow-up (Fig. 11.6): Progression to CMML                 21 × 109/L. Immunophenotype of CD34+ blasts: positive for
   During the following 5 years, progressive leukocytosis with        CD13, CD200; partially positive for CD117, HLA-DR,
monocytosis and splenomegaly developed. The CPC was:                  CD38, CD11c, MPO. FISH analysis of bone marrow cells
WBC 30.64 × 109/L (19% monocytes, blast cells 1%), hemo-              revealed a monosomy 7 (Fig. 11.7). The core biopsy showed
globin 9.8 g/dL, platelet count 68 × 109/L, LDH 970 U/L.              a diffuse infiltration by CD33- and CD34-positive blasts
   Molecular testing by NGS (targeted sequencing)                     associated with a dense reticulin meshwork consistent with
revealed mutations in the SRSF2-gene (ex1), the TET2                  fibrosis grade 2.
a b
                                                            MGG
                                                            MGG
                                                            MG                                                                       MGG
Fig. 11.5 CMML initially presenting as MDS. (a) Peripheral blood      and myeloid dysplasia including the presence of pseudo-Pelger cells
smear with anisocytosis of red blood cells and relative monocytosis   (d) (MGG). (e, f) Hypercellular core biopsy showing dysplasia of
insufficient for the diagnosis of CMML (MGG). (b–d) Bone marrow       megakaryocytes, diminished erythropoiesis, left-shifted granulopoiesis,
aspirate demonstrating megakaryocytic dysplasia including the pres-   and monocytosis (Giemsa). (g, h) Focal increase in monocyte precur-
ence of micromegakaryocytes (c), monocyte precursors and monocytes,   sors that are predominantly negative for NASDCL
11   Myelodysplastic/Myeloproliferative Neoplasms (MDS/MPN)            321
c d
                                                        MGG         MGG
                                                                    MG
                                                                     GG
e f
Giemsa Giemsa
g h
                                                   NA
                                                   NAS
                                                     SD
                                                      DC
                                                       CL
                                                        L
                                                   NASDCL         NASDCL
                                                                  NA
                                                                  NAS
                                                                    SD
                                                                     DCCL
                                                                        L
a b
                                                          Giiem
                                                          G   ms
                                                               saa                                                                 Giemsa
c d
                                                          Giie
                                                          G  em
                                                              msa
                                                              msa
                                                               sa                                                                  Giemsa
                                                                                                                                   Giie
                                                                                                                                   G  emm
                                                                                                                                        msa
                                                                                                                                         sa
                                                                                                                                         sa
e f
CD61 CD14
Fig. 11.6 Overt CMML, JAK2 V617F positive. (a–d) The Giemsa-            lineage is immunolabeled by CD14 antibody. (d, g) Note intertrabecu-
stained bone marrow core biopsy is completely devoid of fat cells as a   lar nodules composed of mononuclear cells that correspond to (g)
consequence of myelomonocytic proliferation. (d, e) Abundant mega-       CD123-positive cells derived from the monocytic lineage. (h)
karyocytes focally form loose clusters of large CD61-positive polymor-   Predominant grade 1 reticulin fibrosis with focal increase to grade 2
phous cells with features of MPN rather than MDS. (f) The monocytic      present in less than 30% of bone marrow spaces
11       Myelodysplastic/Myeloproliferative Neoplasms (MDS/MPN)                                                                             323
g h
CD123 GOMORI
a b
Giemsa Giemsa
c d
CD34 CD33
Fig. 11.7 Transformation of the CMML in AML MRC. (a, b) The             cells are rarely observed. (f) Megakaryopoiesis is strikingly diminished
Giemsa-stained core biopsy reveals abundant blasts showing a pale       with the exception of scattered CD61-positive micromegakaryocytes
basophilic cytoplasm, nuclei with round or slightly folded nuclei and   and megakaryoblasts. (g, h) The reticulin fiber network is increased to
expression of (c) CD34 and (d) CD33. (e) CD14-positive monocytic        grade 2 with small foci of grade 3 fibrosis
324                                  11   Myelodysplastic/Myeloproliferative Neoplasms (MDS/MPN)
e f
CD14 CD61
g h
GOMORI GOMORI
a b
                                                               HE
                                                               HE                                                                        HE
c d
                                                               HE                                                                        HE
                                                                                                                                         HE
Fig. 11.8 JMML, bone marrow core biopsy. (a–f) H&E stained sec-        separated nuclei forming loose clusters, and (f) a megaloblastoid eryth-
tions showing highly cellular marrow spaces containing abundant        ropoiesis. (g, h) NASDCL+ neutrophil precursors were predominantly
myelomonocytic cells, (c–e) megakaryocytes with single or completely   present in the vicinity of the trabecular bone
11       Myelodysplastic/Myeloproliferative Neoplasms (MDS/MPN)                                                                           327
e f
HE HE
g h
NASDCL NASDCL
a b
HE HE
Fig. 11.9 JMML, skin biopsy, and splenectomy specimen. (a–d)            monocytic cells in the red pulp adjacent to lymphoid components of the
Predominantly perivascular infiltrates of the skin by JMML cells with   white pulp is evident on H&E stained sections of the spleen
(d) a high content of CD14+ monocytes. (e–h) Expansion of myelo-
328                              11   Myelodysplastic/Myeloproliferative Neoplasms (MDS/MPN)
c d
HE CD14
e f
HE HE
g h
                        HE
                        HE                                                             HE
                                                                                       HE
                                                                    11.3.6.2 Osteopetrosis
11.3.5 Genetics                                                    Interestingly, malignant infantile osteopetrosis (IMO)—a
                                                                    genetic disorder of bone resorption—may be misdiagnosed
The majority of cases have a normal karyotype while mono-           as JMML ([19]; Fig. 11.10). Genetic analyses are helpful to
somy 7 is detected in about one third of patients and a minor-      exclude rare cases IMO that is characterized by mutations in
ity harbors other aberrations [14].                                 TCIRG1, CLCN7, SNX210, OSTM1, or rarely, in RANK and
    Approximately 90% of patients with JMML harbor                  RANKL [19].
largely mutually exclusive mutations in PTPN11, NF1,
NRAS, KRAS, or CBL in their leukemic cells resulting in
hyperactivation of the RAS-MAPK pathway [17].
a b
                                                            MG
                                                            M GG                                                                 MGG
c d
                                                         Giiem
                                                         G   mssa
                                                                a                                                             Giem
                                                                                                                              Gi
                                                                                                                              Giemsa
                                                                                                                                 em
                                                                                                                                 emsa
                                                                                                                                  msa
                                                                                                                                   sa
Fig. 11.10 Malignant infantile osteopetrosis. (a) Increased white   shifted myelopoiesis with dysplastic features, monocytosis, decreased
blood cells with evidence of monocytosis, presence of myelocytes,   erythropoiesis, and dysplastic megakaryocytes with the presence of
metamyelocytes and normoblasts (peripheral blood smear, MGG). (b)   micromegakaryocytes (b, MGG; c–g, Giemsa; h, NASDCL)
Bone marrow aspirate and (c–h) core biopsy showing increased left
330                                                             11   Myelodysplastic/Myeloproliferative Neoplasms (MDS/MPN)
e f
Giemsa Giemsa
g h
Giemsa NASDCL
Case History                                                                   pseudo-Pelger-Huet cells (Fig. 11.11). The bone marrow core
A 71-year-old lady was admitted with weakness, malaise, and                    biopsy was hypercellular with a predominance of the granulo-
splenomegaly. CBC at presentation: WBC 98 × 109/L, hemo-                       cytic lineage exhibiting features of dysgranulopoiesis. The
globin 7.9 g/dL, MCV 94 fL, platelet count 68 × 109/L. The                     erythroid lineage was diminished. Megakaryocytes were gen-
differential count was as follows: blasts 2%, promyelocytes                    erally small and dysplastic. CD34+ blasts accounted for 9% of
10%, myelocytes 8%, metamyelocytes 13%, band forms 27%,                        hematopoietic cells. By FISH, no karyotypic abnormalities
neutrophils 25%, eosinophils 4%, basophils 1%, monocytes                       were found. Molecular analyses revealed SETBP1, AXL1,
3%, lymphocytes 7%. Cytologic examination of blood smears                      FLT3 ITD, and CBL mutations. No BCR-ABL1 or JAK2, MPL,
showed abundant neutrophilic precursors and abnormalities of                   or calreticulin mutations were found. These findings were
neutrophils including hypolobulation, hypergranularity, and                    consistent with a diagnosis of aCML.
a b
MGG MGG
c d
                                                                    MGG                                                                           MGG
                                                                                                                                                  MG
                                                                                                                                                  MGG
Fig. 11.11 Atypical chronic myeloid leukemia, BCR-ABL1-negative. (a–           The hypercellular bone marrow core biopsy exhibits granulocytic prolifera-
d, MGG) Peripheral blood smears showing neutrophilic precursors and            tion with maturation but left shift and dysplasia (e, f, Giemsa; g, NASDCL).
dysplastic hypolobulated neutrophils. Note the presence of rare blasts in d.   (h) CD34+ blasts are increased but represent <10% of hematopoietic cells
11   Myelodysplastic/Myeloproliferative Neoplasms (MDS/MPN)           333
e f
Giemsa Giemsa
g h
NASDCL CD34
Case History                                                              MPN-like cells, and formed loose clusters. The erythropoi-
A 74-year-old male patient was diagnosed with anemia, leu-                esis was decreased and showed rare islands with disturbed
kocytosis, thrombocytosis, and mild splenomegaly. CBC at                  maturation. Granulopoiesis was increased and left shifted.
presentation: 16.1 × 109/L, hemoglobin 8.9 g/dL, MCV                      Blasts accounted for 4% of nucleated bone marrow cells.
96 fL, platelet count 470 × 109/L. The differential count was             Karyotyping showed a monosomy 7. Genetic studies by
as follows: blasts 0%, band forms 4%, neutrophils 48%,                    NGS revealed TET2 and AXL1 mutations but no BCR-ABL1
eosinophils 1%, monocytes 4%, lymphocytes 41%.                            fusion gene and no JAK2, PDGFRA, PDGFRB, FGFR1,
   The bone marrow was hypercellular (Fig. 11.12).                        PCM1-JAK2 rearrangements. Since no criteria of any other
Megakaryocytes were increased, showed MDS- and MPN-                      MDS/MPN were present, the diagnosis was MDS/
like features with a mixture of small dysplastic and large                MPN-U.
a b
                                                               MGG
                                                               MG                                                                      MGG
c d
                                                               MGG
                                                               MG                                                                   Giemsa
                                                                                                                                    Gi
                                                                                                                                    G ie
                                                                                                                                       em
                                                                                                                                        ms
                                                                                                                                         sa
                                                                                                                                          a
Fig. 11.12 Myelodysplastic/myeloproliferative neoplasm, unclassifi-       hypercellularity (d, Giemsa). Erythroid islands are rare but enlarged
able (MDS/MPN-U). a–c (MGG) bone marrow aspirate smears show-             with decreased maturation (e, Giemsa, f, CD71). Both small hypolobu-
ing clusters of small dysplastic and large megakaryocytes, left-shifted   lated and enlarged megakaryocytes are present (g, CD61). No reticulin
myeloid cells with hypogranulated and hypolobated forms and dimin-        fibrosis (h, Gomori)
ished erythropoiesis. Examination of bone marrow core biopsy reveals
336                                   11   Myelodysplastic/Myeloproliferative Neoplasms (MDS/MPN)
e f
Giemsa CD71
g h
                          CD6
                          CD61
                          CD611                                                        Gomori
                                                                                       G
                                                                                       Goomo
                                                                                          mori
                                                                                             r
MDS/MPN-RS-T
a b
                                                         Giiem
                                                         Giemsa
                                                            ems
                                                            em sa
                                                                a                                                                Giemsa
c d
                                                              Iron                                                                    IIron
                                                                                                                                      Irro
                                                                                                                                         onn
Fig. 11.13 (a, b; MGG) Aspirate smears showing cluster of large        megakaryocytes with a predominance of large hyperlobulated cells but
MPN-type and small dysplastic more MDS-type megakaryocytes. (c, d;     also small dysplastic forms. The erythroid series represents about 60%
Prussian blue stain) The presence of abundant ring sideroblasts is     of nucleated cells and shows megaloblastoid features and a decreased
detected by iron stain performed on bone marrow aspirate smears. The   maturation (e, f, Giemsa; g, CD61; h, CD71)
hypercellular bone marrow core biopsy contains markedly atypical
11       Myelodysplastic/Myeloproliferative Neoplasms (MDS/MPN)          339
e f
HE HE
g h
                                                           CD
                                                           CD71
                                                             71
                                                           CD71        CD61
                                                                       C
                                                                       CD
                                                                        D6
                                                                         61
                                                                          1
25. Gotlib J, Maxson JE, George TI, Tyner JW. The new genetics of                blasts and thrombocytosis. In: Swerdlow SH, Campo E, Harris NL,
    chronic neutrophilic leukemia and atypical CML: implications for             Jaffe ES, Pileri SA, Stein H, Thiele J, editors. WHO classification
    diagnosis and treatment. Blood. 2013;122(10):1707–11.                        of tumours of haematopoietic and lymphoid tissues. Lyon: IARC;
26. Wang SA, Hasserjian RP, Fox PS, et al. Atypical chronic myeloid              2017. p. 93–4.
    leukemia is clinically distinct from unclassifiable myelodysplastic/   32.   Malcovati L, Cazzola M. Recent advances in the understanding of
    myeloproliferative neoplasms. Blood. 2014;123(17):2645–51.                   myelodysplastic syndromes with ring sideroblasts. Br J Haematol.
27. Linder K, Iragavarapu C, Liu D. SETBP1 mutations as a bio-                   2016;174:847–58.
    marker for myelodysplasia /myeloproliferative neoplasm over-           33.   Schmitt-Graeff AH, Teo SS, Olschewski M, Schaub F, Haxelmans
    lap syndrome. Biomark Res. 2017;5:33. https://doi.org/10.1186/               S, Kirn A, Reinecke P, Germing U, Skoda RC. JAK2V617F muta-
    s40364-017-0113-8.                                                           tion status identifies subtypes of refractory anemia with ringed sid-
28. Orazi A, Bennett JM, Bain BJ, Baumann I, Thiele J, Bueso-Ramos               eroblasts associated with marked thrombocytosis. Haematologica.
    C, Malcavoti L. Myelodysplastic/myeloproliferative neoplasm,                 2008;93:34–40.
    unclassifiable. In: Swerdlow SH, Campo E, Harris NL, Jaffe ES,         34.   Patnaik MM, Tefferi A. Refractory anemia with ring sideroblasts
    Pileri SA, Stein H, Thiele J, editors. WHO classification of tumours         (RARS) and RARS with thrombocytosis (RARS-T)—“2017 update
    of haematopoietic and lymphoid tissues. Lyon: IARC; 2017.                    on diagnosis, risk-stratification, and management”. Am J Hematol.
    p. 95–6.                                                                     2017;92(3):297–310.
29. Orazi A, Germing U. The myelodysplastic/myeloproliferative             35.   Deininger WN, Jeffrey W, Tyner JW, Solary E. Turning the tide in
    neoplasms: myeloproliferative diseases with dysplastic features.             myelodysplastic/ myeloproliferative neoplasms. Nat Rev Cancer.
    Leukemia. 2008;22(7):1308–19.                                                2017;17:425–40.
30. Meggendorfer M, Bacher U, Alpermann T, Haferlach C, Kern               36.   Schmitt-Graeff A, Thiele J, Zuk I, Kvasnicka HM. Essential throm-
    W, GambacortiPasserini C, Haferlach T, Schnittger S. SETBP1                  bocythemia with ringed sideroblasts: a heterogeneous spectrum of
    mutations occur in 9% of MDS/MPN and in 4% of MPN cases                      diseases, but not a distinct entity. Haematologica. 2002;87(4):392–9.
    and are strongly associated with atypical CML, monosomy 7, iso-        37.   Cazzola M, Rossi M, Malcovati L. Biologic and clinical signifi-
    chromosome i(17)(q10), ASXL1 and CBL mutations. Leukemia.                    cance of somatic mutations of SF3B1 in myeloid and lymphoid
    2013;27(9):1852–60.                                                          neoplasms. Blood. 2013;121:260–9.
31. Orazi A, Hasserjian RP, Cazzola M, Thiele J, Malcavoti L.
    Myelodysplastic/myeloproliferative neoplasm with ring sidero-
                                 Myelodysplastic Syndromes (MDS)
                                                                                                                                            12
Table 12.2 Myelodysplastic syndromes: the spectrum of common and           reticulocyte counts. Patients with an absolute neutrophil
heterogeneous featuresa                                                    count <1.8 × 109/L are considered to be neutropenic.
    Common features                                                        Thrombocytopenia is diagnosed when thrombocytes are
    – Ineffective hematopoiesis with one or more peripheral cytopenias     <100 × 109/La [2]. Anemia is the most frequent symptom of
    – Morphologic dysplasia of maturing hematopoietic elements
                                                                           MDS, while isolated neutropenia or thrombocytopenia are
    Heterogeneous features
    – Clinical course: indolent with a nearly normal life expectancy to   rare [4]. Death may result from the consequences of refrac-
        rapidly progressing to AML                                         tory cytopenias or from transformation into AML with MDS-
    – Different numbers of cytopenias                                      related changes (AML MRC). The clinical course is highly
    – Variable expansion of myeloblasts (<20%)                             variable ranging from a rather indolent disease in low-risk
    – Different cytogenetic and molecular abnormalities                    MDS to a rapidly unfavorable outcome in high-risk disease.
    – Variable risk for shortened overall and leukemia-free survival
                                                                           Predicting the individual risk category is important for
        (see prognostic scoring systems)
    Exceptions                                                             patient counseling and treatment recommendations.
    • AML- defining cytogenetic features without consideration of         Prognostic factors in MDS can be divided into patient-related
       blasts cell percentage                                              or disease-related factors. Patient-related factors can include
     Cytogenetic abnormalities typical for de novo AML                    age at diagnosis, performance status, and comorbidities.
     t(8;21)(q22;q22)RUNX1-RUNX1T1, inv(16)(p13.1q22), t(16;16)           Disease-related factors include pathological, laboratory, and
      (p13.1;q22)CBFB-MYH11, t(15;17)(q24.1;q21.2); PML-RARA
                                                                           biological features [12]. Different prognostic scoring systems
    • MDS secondary to chemotherapy or irradiation: Therapy-
       related myeloid neoplasms                                           such as the International Prognostic scoring system (IPSS),
Modified according to [2, 7]
a                                                                          the revised IPSS (IPSS-R), the World Health Organization
                                                                           (WHO) Prognostic Scoring System (WPSS), and others are
                                                                           currently used to risk-stratify MDS patients. These systems
immunophenotyping, and the evaluation of the genetic and                   are principally based on the karyotype, the bone marrow
epigenetic profile by established and new technologies.                    blast percentage, and/or cytopenias, transfusion dependency,
                                                                           and/or features of the WHO classification but have different
                                                                           cutoffs for various parameters [12].
12.2         Epidemiology
12.4.1 Special Stains in MDS                                     ated by morphologic analysis of smears and biopsies are pro-
                                                                  vided in Table 12.4.
The microscopic analyses of smears prepared from peripheral
blood and bone marrow aspirates as well as histologic slides      12.4.2.1 The Peripheral Blood in MDS
obtained from bone marrow core biopsies have still an impor-      In peripheral blood smears, red blood cells frequently show
tant impact on the work-up of MDS particularly as a rapid         anisocytosis, poikilocytosis and macrocytosis. Dual hyper-
initial diagnostic procedure [14]. A high technical quality of    and normochromic cell populations can be observed. In the
specimens is mandatory [15]. Basic staining methods for the       rare cases of acquired α-thalassemia-MDS (ATMDS), hypo-
evaluation of aspirates include the panoptic Pappenheim’s         chromasia and microcytosis are present. Granulocytes of
(May Grünwald-Giemsa, MGG) or Wright’s stain, Pearl’s (or         MDS patients are generally scarce reflecting granulocytope-
Berlin blue) iron stain and, as required, the cytochemical        nia. Cytoplasmic hypogranularity or agranularity are often
determinations of myeloperoxidase (MPO), and non-specific         noted. Hyposegmentation of nuclei may be similar to the
esterase, e.g., α-naphthyl acetate esterase. Bone marrow core     inherited Pelger-Huet anomaly (so-called pseudo-Pelger
biopsies should preferentially be fixed in buffered 4% forma-     cells). In addition, hypersegmented or giant forms of nuclear
lin, decalcified in EDTA and paraffin-embedded. In addition      rings may be present. Abnormalities of platelets include
to panoptic stains (hematoxylin & eosin and/or Giemsa), spe-      anisocytosis, hypo- or hypergranularity, and giant forms.
cial stains such as naphthol-ASD-       chloroacetate esterase   Platelet counts are often decreased with the exceptions of
(NASDCL) for granulopoiesis, Pearl’s for hemosiderin,             MDS associated with isolated del(5q) or 3q26 abnormalities.
Gomori’s silver impregnation for reticulin fibers, and tri-       Patients with these subsets of MDS have often normal or
chrome stains in order to demonstrate a dense collagen mesh-      increased platelets. In MDS-EB-1 or -2, the peripheral blasts
work should be applied to sections cut from core biopsies. A      represent 2–4% or 5–19%, respectively, and may contain
wide panel of antibodies is now available for further evalua-     Auer rods in MDS-EB-2. The blast cell count should include
tion of the different myeloid cell lineages, highlighting the     myeloblasts but also monoblasts, promonocytes, and mega-
presence of blasts, detecting lymphoid infiltrates, cancer        karyoblasts but no proerythroblasts [14].
cells, and characterizing stromal components in bone marrow
core biopsies. Immunophenotyping of the neoplastic popula-        12.4.2.2      he Bone Marrow in MDS: Dysplasia
                                                                               T
tion by flow cytometry (FC) is essential for the diagnostic                    Involving the Hematopoietic Lineages
work-up and monitoring of AML and is increasingly recog-          According to the 2008 WHO criteria, unequivocal dysplastic
nized as part of an integrated diagnosis in MDS [16]. However,    features in ≥10% of cells of at least one of the myeloid lin-
conventional stains and immunolabeling techniques applied         eages are required to establish the diagnosis of MDS [2].
to cytologic and histologic specimens can still significantly     Exceptions are cases with MDS-U (see Table 12.1).
contribute to the characterization of these MNs.                  Currently, dysplastic features of ≥10% of precursors of at
Immunohistochemistry of core biopsies remains most useful         least two myeloid lineages are required for the diagnosis of
in cases presenting with hypoplastic and/or fibrotic variants     MDS-MLD.
of MDS and AML in which sufficient material for immuno-              Erythroid lineage: Erythropoiesis is often, but not in
phenotyping by FC is not available. Moreover, FISH to detect      every MDS case, the preponderant lineage. Enlarged ery-
rearrangements and losses or gains of genetic material can be     throid islands containing increased numbers of immature
routinely performed on smears, as well as on sections cut         precursors may be dislocated from the center of the marrow
from paraffin-embedded biopsies [14].                             spaces to the niches adjacent to the trabecular bone. Erythroid
                                                                  precursors tend to show abnormalities such as megaloblas-
                                                                  toid nuclear changes, nuclear lobulation, multinuclearity,
12.4.2 D
        ysplastic Features Observed                              internuclear chromatin bridges, cytoplasmic vacuolization,
       in Peripheral Blood and Bone Marrow                        or in rare cases PAS-positivity. While the latter features may
       Specimens                                                  be detected on cytologic bone marrow aspirate smears, as
                                                                  well as on sections from core biopsies, the presence of ring
The generally hyper-, rarely hypocellular, and occasionally       sideroblasts is revealed only on Prussian blue stained bone
fibrotic bone marrow shows dysplastic features in ≥10% of         marrow smears. Ring sideroblasts are erythroid precursors
cells of at least one of the hematopoietic lineages. Rare         containing ≥5 iron granules which encircle the nucleus and
exceptions are cases with dysplastic features in <10% of          correspond to mitochondrial iron deposits. Erythropoiesis
myeloid cells, but harboring MDS-type cytogenetic altera-         can be immunolabeled by antibodies to hemoglobin, gly-
tions. These cases are assigned to as MDS-U (see Table 12.1).     cophorin C, or CD71. Abnormal erythroid precursors may
Features that are suggestive of an MDS and may be evalu-          also express CD117.
346                                                                                                   12   Myelodysplastic Syndromes (MDS)
Table 12.4 Morphologic and immunohistochemical features of dysplasia in peripheral blood and bone marrowa
Hematopoietic         Cytology of bone marrow aspirate        Cytology of peripheral   Histology and immunohistochemistry of bone marrow
lineage               smears                                  blood smears             Bone marrow core biopsies
Erythropoiesis/red    Megaloblastic changes                   Anisopoikilocytosis      Maturation defects
blood cells           Maturation defects                      Hyperchromasia           Makroblastic hyperplasia
                      Nuclear lobulation, budding             Dual RBC populations     Enlarged erythroid island
                      Bi-/multinuclearity                     Siderocytes              Abnormal paratrabecular localization
                      Internuclear chromatin strands          Basophilic stippling     of erythropoiesis
                      Abnormal mitoses                                                 IH: glycophorin C, hemoglobin, CD71
                      Ring sideroblasts
                      PAS+ cytoplasmic inclusions
                      Cytoplasmic vacuolization
Granulopoiesis/       Maturation defects                      Pseudo-Pelger cells      Maturation defects
neutrophils           Hypo-/hypergranularity                  Hypersegmentation        Increased number of immature precursors
                      Abnormal granules                       Hypogranularity          Abnormal localization of immature precursors
                      Myeloperoxidase deficiency              Giant forms              (ALIP) in the central marrow cavity
                                                              Ring nuclei              IH: myeloperoxidase, CD13, elastase, CD34
Megakaryopoiesis/     Micromegakaryocytes                     Anisocytosis             Micromegakaryocytes
platelets             Large cells                             Giant platelets          Nuclear abnormalities (mono-/multilobation)
                      Monolobated or multilobated forms       Vacuolization            Megakaryocyte clustering
                      Hypersegmentation                                                IH: CD61, CD42b, CD41 (CD34 in some cases)
                      Nuclear-cytoplasmic asynchrony
Blast cells           Percentage of blasts                    Percentage of blasts     Percentage of blasts
                      Auer rods                               Auer rods                Spatial distribution of blasts (disseminated vs.
Hypergranularity      Agranularity/hyper-granularity          A-/hypergranularity      clusters)
                      Myeloperoxidase                         Cytochemical features    Immunohistochemical characterization:
                                                              (myeloperoxidase-/       CD34, CD117, CD33
                                                              esterase positivity)     (CD14, myeloperoxidase, CD61,CD42b)
Additional features   Percentage of dysplastic cells in the                            Bone marrow architecture
                      different myeloid lineages                                       Cellularity
                      Lymphocytes, plasma cells,                                       Fibrosis
                      Monocytic proliferation                                          Vascularity
                      Hemosiderin in macrophages                                       Lymphocytes, plasma cells
                                                                                       Monocytic proliferation
                                                                                       Hemosiderin in macrophages
                                                                                       IH: p53expression
IH immunohistochemistry, RBC red blood cells
a
 Modified from [14]
   Granulocytic lineage: Granulopoiesis is often hypo-                    from micromegakaryocytes to small mononuclear, hypo-
blastic and left-shifted and shows maturation defects.                    or bilobed megakaryocytes, cells with completely and
Abnormal hypo-or hypergranularity including pseudo-Che-                   widely separated nuclei or large hyperlobated forms.
diak-Higashi granules may be present. Similar to blood                    Megakaryocytes of smaller than normal size showing non-
smears, hyposegmented pseudo-Pelger-Huet cells may be                     lobated round nuclei are typical for MDS del(5q). Myeloid
seen. Granulation defects are evident especially on aspirate              neoplasm harboring inv(3)/t(3;3)(q21.3q26.2) or t(3;3)
smears, while the bone marrow core biopsy may reveal the                  (q21.3q26.2) GATA2, MECOM that may present as MDS
atypical localization of immature CD34+ precursors                        or AML according to the blast cell count megakaryocytes
(ALIP) in the central parts of the marrow spaces.                         are small and typically mono- or bilobed. The immuno-
Immunohistochemistry for the detection of granulopoietic                  phenotype of megakaryocytes is characterized by the
cells include antibodies to myeloperoxidase, elastase,                    expression of CD42b, CD41 or CD61 and positive for
CD13, and CD33.                                                           linker of activated T lymphocyte and factor VIII–related
   Megakaryocytic lineage: Megakaryopoiesis may be                        antigen. In MDS, megakaryocyte precursors may also be
hyperplastic, particularly in MDS with isolated del(5q) or                immunolabeled by CD34 antibodies.
inv(3)(q21q26)/t(3;3)(q21;q26) but may also be dimin-                        Blasts: On bone marrow aspirate smears, blast cells
ished. On slides obtained from core biopsies, loose clus-                 show cytological features similar to those observed in the
ters of polymorphic cells may be observed and atypically                  peripheral blood. Megakaryoblasts, monoblasts, and pro-
localized in the periphery of the bone marrow spaces.                     monocytes have to be included when the percentage of
Megakaryocyte size and shape are quite variable ranging                   blasts is evaluated. The presence of myeloid blasts and
12   Myelodysplastic Syndromes (MDS)                                                                                          347
immature myeloid precursors may be highlighted by CD34              moderate to marked fibrosis grade 2 and 3 characterized by a
and CD117 immunohistochemistry.                                     diffuse dense increase in reticulin and collagen fibers which
                                                                    may even be associated with osteosclerosis is an important
12.4.2.3        he Bone Marrow Core Biopsy in MDS:
               T                                                    issue in MDS and has to be mentioned in each pathology
               Bone Marrow Architecture                             report. Grading of fibrosis according to the content of so-
               and Stromal Components                               called reticulin and collagen fibers should be performed [18].
The histologic examination of BMTBs is included in the cur-         A hyperfibrotic MDS (MDS-F) is reported in about 10–20%
rent diagnostic approach of MDS to assess marrow cellular-          of all MDS cases. It has been shown that a relevant moderate
ity, fibrosis, and topography. In addition to dysplastic features   to marked marrow fibrosis was associated with multilineage
of the myeloid lineages, morphologic alterations in MDS             dysplasia, high transfusion requirement, and poor-risk cyto-
pertain to topographical organization, cellularity, microves-       genetics [19].
sel density, fiber content, and immune and stromal cells. The           Hemosiderin: MDS patients frequently receive transfu-
microenvironment comprises among others macrophages,                sions that can lead to iron overload. Iron-induced toxicity has
endothelial cells, fibroblasts/myofibroblasts, osteoblasts,         a negative effect on hematopoietic stem cells and may con-
osteoclasts, adipocytes, extracellular matrix components,           tribute to MDS progression [20]. The amount of iron storage
adhesion molecules, soluble factors, and other components           by bone marrow macrophages can be evaluated both on aspi-
of the niche. The mesenchymal niche is actually considered          rate smears and on Pearl’s stained sections of core biopsies.
as a critical contributor to disease initiation and evolution       Hemosiderin granules in erythroblasts, especially in ring
[15]. In the normal bone marrow, precursor cells of the three       sideroblasts, are detected in bone marrow aspirate smears
myeloid lineages reside in specialized bone marrow com-             and not in sections cut from paraffin-embedded bone marrow
partments where they survive, differentiate, and give rise to       core biopsies.
mature blood cells.                                                     Microvessels: MDS is often accompanied by an increase
    Topography: In MDS spatial disturbances are frequently          in microvascular structures that may be visualized by CD34
observed. Precursors of the three myeloid lineages are often        immunohistochemistry.
dispersed throughout the marrow spaces. Collections of                  Immune cells: An immune-mediated impairment of
erythropoietic cells and megakaryocytes are no longer con-          hematopoietic stem and progenitor cells, increased apopto-
fined to the central areas close to marrow sinuses but may be       sis, high cytokine levels, and changes in the hematopoiesis-
dislocated to the paratrabecular zones. By contrast, granulo-       supporting microenvironment may contribute to the
poietic precursors which are normally localized in the para-        pathogenesis especially of hypoplastic MDS. Decreased
trabecular niche may be found in the central parts of marrow        CD8 + T cells, less IFN-γ secretion by CD8 + T cells,
spaces. An abnormal localization of clusters (3–5 cells) or         increased exhausted TIM3 + CD8 + T cells with lower
aggregates (>5 cells) of myeloblasts and promyelocytes              perforin and granzyme B expression and higher CD95
(ALIP) observed in the central marrow cavity of BMTB sec-           expression have been detected in MDS [21]. It has been sug-
tions may be observed. The presence of several ALIPs as well        gested that the expansion of mutated clones may be driven
as CD34+ cell aggregates has been shown to be associated            by inflammation in some patients with lower-risk MDS.
with an increased risk of leukemic transformation in MDS.
    Bone marrow cellularity: Since the cellularity may vary
between different marrow areas, the overall cellularity may         12.5     Genetics in MDS
more accurately be assessed on BMTBs than on smears. In
MDS, cellularity is often increased but may also be normal          12.5.1 Chromosomal Abnormalities
or even decreased. The hypoplastic variant of MDS which is
observed in about 10% of all MDS cases may be diagnosti-            Numerical and structural chromosomal abnormalities are
cally challenging. Depending on the age of the patient, an          present in about 50% of MDS cases. Cytogenetic aberrations
MDS should be considered as hypoplastic MDS when fat                can be detected by conventional chromosome banding analy-
cells occupy about 70–80% of the bone marrow spaces. The            sis of bone marrow metaphases or by fluorescence in situ
differential diagnosis includes idiopathic cytopenia of uncer-      hybridization (FISH) of bone marrow cells and circulating
tain clinical significance (ICUS), aplastic anemia, and toxic       CD34+ cells [22]. In contrast to AML, balanced chromo-
or immunologic bone marrow damage. A detailed search for            somal translocations are rare in MDS while unbalanced
dysplastic features is warranted and may allow a discrimina-        translocations are more frequent. Losses or gains of large
tion between aplastic anemia and hypoplastic MDS or refrac-         segments of chromosomes include −7/del(7q), −5/del(5q),
tory cytopenia of childhood [17].                                   +8, dup(1q), del(20q), del(11q), del(12p)/t(12p), del(17p)/
    Fibrosis: A mild increase in bone marrow reticulin fiber        iso(17q), del(18q), +21q gains, del(13q), der(1;7)(q10;p10).
content grade 1 is of no prognostic relevance in MDS. However,      Complex karyotypes harboring ≥3 chromosomal aberrations
348                                                                                                12   Myelodysplastic Syndromes (MDS)
are frequently associated with TP53 mutation and a dismal          Table 12.5 Selected driver genes involved in MDSa
prognosis.                                                             Function/pathway     Mutated genes, impact on outcome
   Cytogenetic aberrations are associated with different               RNA splicing         SF3B1 (favorableb), SRSF2 (adverseb), U2AF1
prognoses and represent important components of the prog-                                   (adverseb), U2AF2, SF1
                                                                       DNA methylation      TET2 (neutral), DNMT3A (neutral), IDH1/
nostic scoring systems. For example, cytogenetic subgroups
                                                                                            IDH2 (neutral)
defined according to the IPSS-R are: very good: −Y, del(11q);          Chromatin            ASXL1 (adverseb), EZH2 (adverse), MLL2
good: normal, del(5q), del(12p), del(20q), double including            modification
del(5q); intermediate: del(7q), +8, +19, i(17q), any other             Transcription        RUNX1 (adverse), ETV6, GATA2, CEBPA,
single or double independent clones; poor: −7, inv(3)/t(3q)/           regulation           CUX1, BCOR
                                                                       Checkpoint/cell      TP53 (adverse), CDKN2A
del(3q), double including −7/del(7q); complex: three abnor-
                                                                       cycle
malities; very poor: complex: >3 abnormalities [23].                   DNA repair           ATM, BRCC3, FANCL
   In    addition,    copy-neutral    loss-of-heterozygosity           RAS signaling        PTPN11, NF1, NRAS, KRAS, CBL
(CN-LOH) or uniparental disomy (UPD), which are com-                   Cohesin complex      STAG2, RAD21
monly seen in chromosomes 1p, 4q, 7q, 11q, 13q, 14q, 17p               Other signaling      JAK2, KIT, MPL, FLT3
may be detected in MDS. Chromosomal abnormalities are                  molecules
no stable features but may increase or even disappear during       Modified from [12, 23]
                                                                   a
                                                                   blasts <5%
                                                                   b
the disease course.
Case History 1. Myelodysplastic Syndrome with Ring                        35% ring sideroblasts. No dysplastic features of the myeloid
Sideroblasts and Single Lineage Dysplasia (MDS-RS-                       or megakaryocytic lineages were observed. The bone core
SLD) (Fig. 12.1)                                                          biopsy also showed a predominance of partially macroblastic
A 59-year-old woman was diagnosed with macrocytic ane-                    erythroid precursors forming large clusters. However, as a
mia. The CBC was as follows: WBC 5.8 × 109/L, Hb 8.5 g/                   consequence of the embedding procedure, ring sideroblasts
dL, RBC 2.0 × 1012/L, MCV 105 fL, platelets 153 × 109/L. The              could not be detected in the FFPE material. Mutational anal-
peripheral blood smears showed normal neutrophils and                     ysis revealed a typical SF3B1 mutation. The diagnosis was
platelets but dimorphic erythrocytes. Bone marrow smears                  myelodysplastic syndrome with ring sideroblasts and single
revealed a predominance of the erythroid lineage including                lineage dysplasia (MDS-RS-SLD).
a b
PB PB
c d
Giemsa Giemsa
Fig. 12.1 Myelodysplastic syndrome with ring sideroblasts and single      and macroblastic transformation (c, d; Giemsa; e, f, CD71). (g) Normal
lineage dysplasia (MDS-RS-SL). (a, b) Presence of numerous erythro-       maturation of granulopoiesis (naphthol-ASD-chloroacetate esterase).
blasts showing nuclei encircled by coarse iron granules, a typical fea-   (h) Absence of CD34+ blasts while endothelial cells show a regular
ture of ring sideroblasts on bone marrow smears (Prussian blue stain).    immunoreactivity as positive internal control
The bone marrow core biopsy with evidence of erythroid hyperplasia
12       Myelodysplastic Syndromes (MDS)                  351
e f
CD71 CD71
g h
NASDCL CD34
Case History 2. Myelodysplastic Syndrome with Ring                          dysplastic neutrophils including pseudo-Pelger cells. The
Sideroblasts and Multilineage Dysplasia (MDS-RS-                           bone marrow smears and the core biopsy were hypercellular
MLD) (Fig. 12.2)                                                            and showed a megaloblastoid erythropoiesis, a left-shifted
A 62-year-old male patient was diagnosed with pancytope-                    dysplastic granulopoiesis, and a predominance of small dys-
nia. The CBC was as follows: WBC 3.9 × 109/L, Hb 5.1 g/                     plastic megakaryocytes. CD34+ bone marrow blasts were
dL, RBC 1.82 × 1012/L, MCV 105 fL, MCHC 33 g/dL, plate-                     3%. Molecular analyses revealed a SF3B1 mutation. The
lets 84 × 109/L. The peripheral blood smears showed a dual                  diagnosis was myelodysplastic syndrome with ring sidero-
population of normocytic and macrocytic red blood cells and                 blasts and multilineage dysplasia (MDS-RS-MLD).
a b
                                                                 MGG
                                                                 MG                                                                     M
                                                                                                                                        MG
                                                                                                                                         GG
                                                                                                                                        MGG
c d
                                                                    PB
                                                                    PB                                                                     PB
                                                                                                                                           P B
Fig. 12.2 Myelodysplastic syndrome with ring sideroblasts and multi-        dysplastic myeloid precursors (MGG). (c–e) Evidence of numerous
lineage dysplasia (MDS-RS-MLD). (a) Peripheral blood showing mac-           ring sideroblasts on Prussian blue stained bone marrow smears.
rocytosis associated with increased red cell distribution width. Note the   Strikingly hypercellular bone marrow core biopsy showing trilineage
presence of a dysplastic neutrophil with hypolobated nucleus (MGG).         dysplasia and numerous small partially bilobed megakaryocytes (f, g,
(b) Bone marrow aspirate smear with megaloblastoid erythroblasts and        Giemsa; h, CD61). No excess of blasts was present
12   Myelodysplastic Syndromes (MDS)                     353
e f
PB Giemsa
g h
                                       Gi
                                       Gie
                                         em
                                          mssa
                                       Giemsaa        CD61
                                                      CD
                                                       D61
                                                        61
Case History 3. Myelodysplastic Syndrome with                            were present. The bone marrow aspirate smears showed
Multilineage Dysplasia (MDS-MLD) (Fig. 12.3)                             macroblastic erythropoiesis and prominent granulocytic dys-
A 64-year-old male patient was diagnosed with pancytope-                 plasia with atypically lobated neutrophils. The bone marrow
nia with predominant neutropenia. The CBC was as follows:                core biopsy was mildly hypercellular with evidence of trilin-
WBC 1.8 × 109/L, Hb 11.3 g/dL, RBC 3.24 × 1012/L, MCV                    ear dysplasia and small foci of CD34+ blast cells. Their per-
107 fL, platelets 95 × 109/L. The differential count showed              centage was 3%. The karyotype was normal. Molecular
38% neutrophils, 9% monocytes, 1% eosinophils, and 52%                   analysis revealed TET2 and DNMT3A mutations. The diag-
lymphocytes. Neutrophils frequently showed abnormal                      nosis was myelodysplastic syndrome with multilineage dys-
nuclear lobulation and a hypogranular cytoplasm. No blasts               plasia (MDS-MLD).
a b
                                                               MG
                                                                GG                                                                   MGG
                                                                                                                                     MG
                                                                                                                                     MGG
c d
                                                            Giiem
                                                            G  emssa
                                                                   a                                                              Giemsa
                                                                                                                                  G
                                                                                                                                  Giie
                                                                                                                                     em
                                                                                                                                      ms
                                                                                                                                       sa
                                                                                                                                        a
Fig. 12.3 Myelodysplastic syndrome with multilineage dysplasia           numerous megakaryocytes (Giemsa). (e, f) Left-shifted and diminished
(MDS-MLD). (a) Dysplastic neutrophil, a lymphocyte, and absence of       granulopoiesis surrounding erythroid island and numerous dysplastic
platelets in a peripheral blood smear (MGG). (b) Bone marrow smear       megakaryocytes (naphthol-ASD-chloroacetate esterase). (g) Multiple
showing abundant megaloblastoid erythroid precursors, immature gran-     predominantly small-sized megakaryocytes and some megakaryoblasts
ulocytic cells, and dysplastic neutrophils (MGG). (c, d) Mildly hyper-   immunolabeled by CD61 antibody. (h) Presence of rare small clusters
cellular bone marrow core biopsy with prominent erythropoiesis and       of CD34+ blast cells representing 3% of nucleated cells
12   Myelodysplastic Syndromes (MDS)                      355
e f
                                       N
                                       NA
                                       NASDCL
                                        ASSD
                                           DCCL
                                              L       NASDCL
g h
                                          CD61
                                          CD61          C 34
                                                        CD
                                                        CD34
                                                          3
Case History 4. Myelodysplastic Syndrome with                            The bone marrow core biopsy was hypercellular and showed
Multilineage Dysplasia (MDS-MLD), Monosomy 7, and                        enlarged atypical localized erythropoietic islands replacing
Multiple Gene Mutations (Figs. 12.4 and 12.5)                            granulopoiesis adjacent to the trabecular bone. Granulopoiesis
A 39-year-old woman was admitted with pneumonia. The                     was diminished and left shifted. Multiple small-sized hypolo-
CBC was consistent with pancytopenia: WBC 1.74 × 109/L,                  bated megakaryocytes and monolobated micromegakaryo-
Hb 7.3 g/dL, RBC 1.83 × 1012/L, MCV 106 fL, platelets                    cytes were detected. Karyotyping showed monosomy 7
84 × 109/L. The peripheral blood showed 11% band forms,                  consistent with a poor cytogenetic subgroup.
15% neutrophils, 5% monocytes, and 69% lymphocytes.                         Molecular analysis revealed mutations in the ASXL-1,
Bone marrow aspirate smears showed 48% erythroid precur-                 DNMT3A, EZH2, and PHF6 genes. The diagnosis was MDS-
sors, 4% blasts, 11% lymphocytes, and 37% myeloid cells.                 MLD assigned to the intermediate risk group (IPSS-R 3.5).
a b
                                                              MGG                                                                         MGG
                                                                                                                                          MG
                                                                                                                                           GG
c d
                                                              MGG
                                                                                                                               Giemsa
Fig. 12.4 Myelodysplastic syndrome with multilineage dysplasia           bone marrow core biopsy with increased megalobalstoid erythropoie-
(MDS-MLD), monosomy 7, and multiple gene mutations. (a) Peripheral       sis. Note the abnormal localization of erythropoietic islands close to the
blood smear showing a dysplastic hypolobated neutrophil (MGG). (b,       endosteal bone marrow niche in e (Giemsa). (g, h) Inversion of the ery-
c) Bone marrow aspirate showing megaloblastoid erythroid maturation      throid : myeloid relationship is evident on naphthol-ASD-chloroacetate-
and immature myeloid cells including some blasts. Note the presence of   esterase stained sections
a small bilobed megakaryocyte in c (MGG). (d–f) Mildly hypercellular
12       Myelodysplastic Syndromes (MDS)                                                                                                   357
e f
Giemsa Giemsa
g h
NASDCL NASDCL
a b
CD71 CD71
Fig. 12.5 Myelodysplastic syndrome with multilineage dysplasia         expansion and abnormal localization of the erythroid lineage. (e, f)
(MDS-MLD), monosomy 7, and multiple gene mutations.                    CD61 immunostain to demonstrate the presence of abundant small-
Immunohistochemical stains performed on the biopsy already demon-      sized hypolobated megakaryocytes. (g) Small clusters of CD34+ blasts.
strated in Fig. 12.4. (a–d) Immunolabeling for CD71 highlighting the   (h) Scattered blasts and rare mast cells identified by staining for CD117
358                                 12   Myelodysplastic Syndromes (MDS)
c d
CD71 CD71
e f
CD61 CD61
g h
                        CD
                        CD34
                           34
                        CD34                                   CD117
                                                               C
                                                               CD
                                                                D1
                                                                 11
                                                                  17
Case History 5. Myelodysplastic Syndrome with Excess                      phages but no ring sideroblasts. The bone marrow core
Blasts 1 (MDS-EB-1) (Figs. 12.6 and 12.7)                                 biopsy was moderately hypercellular and showed anomalous
A 66-year-old woman developed pancytopenia. The CBC                       localization of granulopoiesis in the central zones of the bone
was as follows: WBC 3.2 × 109/L, Hb 7.5 g/dL, RBC                         marrow spaces. The erythroid lineage predominantly showed
2.7 × 1012/L, MCV 86 fL, platelets 59 × 109/L. The periph-                immature precursors. There were numerous predominantly
eral blood showed rare dysplastic neutrophils but no blasts.              small-sized dysplastic megakaryocytes. Molecular analysis
The bone marrow aspirate smears showed multilineage dys-                  revealed an unfavorable EZH2 mutation. The diagnosis was
plasia, 9% blasts, and an increased iron content in macro-                myelodysplastic syndrome with excess blasts 1 (MDS-EB-1).
a b
MGG MGG
c d
MGG MPO
Fig. 12.6 Myelodysplastic syndrome with excess blasts-1                   myeloperoxidase). (e, f) Increased iron content of bone marrow macro-
(MDS-EB-1). (a) Hypolobated peripheral blood granulocyte (MGG).           phages but no ring sideroblasts. (g, h) Hypercellular bone marrow core
Bone marrow aspirate smears showing trilineage dysplasia, abnormal        biopsy showing clusters of micromegakaryocytes with non-lobated
bilobed megakaryocytes and small clusters of blast cells (b, c, MGG; d,   nuclei in h (H&E)
360                                                                                                        12   Myelodysplastic Syndromes (MDS)
e f
PB PB
g h
                                                                   H&E
                                                                   H&
                                                                   H&E                                                                        H&E
                                                                                                                                              H
                                                                                                                                              H&
                                                                                                                                               &E
a b
PAS H&E
Fig. 12.7 Myelodysplastic syndrome with excess blasts 1 (MDS-EB-1).          Presence of immature granulocytic precursors in the intratrabecular area
Higher magnification of bone marrow spaces showing immature ery-             is highlighted by naphthol-ASD-chloroacetate-esterase reaction. (e, f)
throid and myeloid precursors and dysplastic megakaryocytes with non-        CD61 immunolabeling demonstrating abundant dysplastic predomi-
lobated or bilobed nuclei. Increased PAS-positive cellular debris in A       nantly small megakaryocytes and some megakaryoblasts. (g, h) Clusters
suggestive of increased intramedullary cell death (a, PAS; b, H&E). (c, d)   of CD34+ blasts accounting for 9% of nucleated cells express CD34
12   Myelodysplastic Syndromes (MDS)                    361
c d
NASDCL NASDCL
e f
CD61 CD61
g h
CD34 CD34
Case History 6. Myelodysplastic Syndrome with Excess                        tially positive for CD13, CD38, MPO, lysozyme, CD33,
Blasts 2 (MDS-EB-2) (Fig. 12.8)                                             CD34 and CD56. The bone marrow core biopsy was hyper-
A 71-year-old male patient was admitted with a suspected                    cellular and showed multilineage dysplasia. Myeloblasts were
diagnosis of acute myeloid leukemia. The CBC was as follow-                 <20% and scattered between hematopoietic precursors or
ing: WBC 5.3 × 109/L, Hb 6.1 g/dL, platelets 43 × 109/L. The                arranged in small groups. The karyotype was complex with
peripheral blood showed rare dysplastic neutrophils and 12%                 >3 cytogenetic abnormalities. Molecular analysis provided
blasts. The bone marrow aspirate smear showed multilineage                  evidence of mutations in ASXL1, RUNX1, and TET2 genes.
dysplasia with rare dysplastic megakaryocytes and 18%                       The diagnosis was myelodysplastic syndrome with excess
blasts. Blasts expressed CD117 and HLA-DR and were par-                     blasts 2 (MDS-EB-2) harboring a high-risk genetic profile.
a b
                                                                 MG
                                                                 M GG
                                                                   GG                                                                      MGG
                                                                                                                                           MG G
c d
NASDCL NASDCL
Fig. 12.8 Myelodysplastic syndrome with excess blasts 2                     ulocytic series (naphthol-ASD-chloroacetate-esterase reaction). (e, f)
(MDS-EB-2). (a, b) Bone marrow aspirate diluted with blood showing          Irregular distribution of the CD71+ erythroid lineage partially arranged
nuclear lobulation of the dysplastic erythroid lineage, granulocytic pre-   in nests partially scattered among myeloid cells. (g, h) Increased
cursors, dysplastic neutrophils, and medium-sized mononuclear blasts        CD34+ blasts representing about 18% of nucleated bone marrow cells
(MGG). (c, d) Dysplastic features and disturbed maturation of the gran-
12   Myelodysplastic Syndromes (MDS)                363
e f
CD71 CD71
g h
CD34 CD34
Case History 7. Myelodysplastic Syndrome with Excess                      Widely separated nuclear lobes were observed in most mega-
Blasts 2 (MDS-EB-2) with Fibrosis Grade 2 (Fig. 12.9)                     karyocytes. Blasts represented about 15% of nucleated cells
A 72-year-old previously healthy male patient was admitted                including the increased erythroid lineage. Gomori’s stain
to the hospital with dyspnea. The CBC was as follows: WBC                 revealed an irregular increased fiber network consistent with
1.9 × 109/L, absolute neutrophil count 0.6 × 109/L, Hb 9.6 g/             reticulin fibrosis grade 2. The karyotype was normal.
dL, RBC 3.19 × 1012/L Hkt 26.6%, MCV 83 fL, platelets                     Molecular analysis identified mutations in DNMT3A, NPM1,
59 × 109/L. The peripheral blood showed 5% erythroblasts                  and RAD21 genes. No JAK2 V617F, MPL, or calreticulin
per 100 nucleated cells and anisopoikilocytosis of red blood              mutations were present. A subsequently performed immuno-
cells. No myeloblasts were detected. The bone marrow aspi-                histochemical stain for NPM1 clearly showed a cytoplasmic
rate resulted in a dry tap. The bone marrow core biopsy was               and nuclear immunoreactivity consistent with the NPM1
markedly hypercellular with less than 5% fat cells. Dysplastic            mutation. The IPSS-R risk score was high [5]. This MDS
features were present in all three hematopoietic lineages.                rapidly transformed into AML-MRC.
a b
                                                            Giemsa                                                                  Giemsa
                                                                                                                                    Giie
                                                                                                                                    G  em
                                                                                                                                        ms
                                                                                                                                         sa
                                                                                                                                          a
c d
                                                                H&E
                                                                H&E
                                                                H&                                                                      H&E
                                                                                                                                        H
                                                                                                                                        H&
                                                                                                                                         &E
Fig. 12.9 Myelodysplastic syndrome with excess blasts 2 (MDS-EB-2)        increase in reticulin fibers consistent with fibrosis grade 2 (Gomori
with fibrosis grade 2. (a–d) Markedly hypercellular bone marrow core      stain). (f) Sheets of immature CD33+ myeloid cells. (g) Immature gran-
biopsy with enlarged atypically localized erythroid islands, incomplete   ulocytic precursors highlighted by naphthol-ASD-chloroacetate-
maturation of myeloid cells, and loose clusters of megakaryocytes         esterase reaction. Note the presence of a large dysplastic megakaryocyte
(Giemsa). (c) Atypical localization of megakaryocytes close to the end-   and nuclear irregularities of erythroid precursors that are negative for
osteal niche. Note the presence of initial osteosclerosis (H&E). (d)      NASDCL. (h) Nuclear and cytoplasmic expression of NPM1 protein as
Megakaryocytes with widely separated nuclei (H&E). (e) Moderate           a morphologic equivalent to of the NPM1 mutation
12   Myelodysplastic Syndromes (MDS)                     365
e f
                                        Go
                                        G
                                        Gomori
                                         ommo
                                            ori
                                              ri       CD33
g h
                                       N
                                       NA
                                        ASSD
                                           DCL
                                            CL
                                       NASDCL          NPM1
Case History 8. Myelodysplastic Syndrome with Isolated                erythroid lineage, and a normal maturation sequence of gran-
del(5q) (Fig. 12.10)                                                  ulocytopoiesis. Blasts represented 1% of nucleated cells. The
A 71-year-old male patient was diagnosed with isolated ane-           hallmark of the bone marrow core biopsy was the presence
mia. The CBC was as follows: WBC 5.4 × 109/L, Hb 9.3 g/               of abundant small-sized hypolobated megakaryocytes.
dL, RBC 3.07 × 1012/L Hkt 27.4%, MCV 103 fL, platelets                Erythropoietic cells were increased and showed partial mac-
143 × 109/L. The peripheral blood showed 52% neutrophils,             roblastosis. Blasts were not increased. The karyotype was
9% monocytes, and 49% lymphocytes. The bone marrow                    46, del(5)(q21q34). Next-generation sequencing showed no
aspirate smear showed numerous small unilobated mega-                 additional mutation. The diagnosis was myelodysplastic syn-
karyocytes, a moderate macroblastic transformation of the             drome with isolated del(5q).
a b
                                                            MG
                                                            M GG                                                                  MGG
                                                                                                                                  M
                                                                                                                                  MGGG
c d
                                                             H&
                                                             H&E                                                                   H&E
                                                                                                                                   H&E
                                                                                                                                   H&
Fig. 12.10 Myelodysplastic syndrome with isolated del(5q). (a, b)     CD61 immunohistochemistry clearly demonstrated the typical features
Bone marrow aspirate smears showing small unilobated megakaryo-       of this MDS type. (g) Presence of enlarged macroblastic CD71+ ery-
cytes as a hallmark of MDS del(5q) (MGG). (c, d) Bone marrow core     throid island. (h) Increased hemosiderin content of bone marrow mac-
biopsy with numerous small hypolabated megakaryocytes (H&E). (e, f)   rophages (Prussian blue stain)
12   Myelodysplastic Syndromes (MDS)                367
e f
CD61 CD61
g h
CD71 PB
Case History 9. Myelodysplastic Syndrome with del(5q),                    with 18% ring sideroblasts. Myeloid blasts were not
Concomitant SF3B1 Mutation, and Ring Sideroblasts                         increased. The bone marrow core biopsy was hypercellular
(Fig. 12.11)                                                              along with increased erythroid precursors and numerous
A 67-year-old woman was diagnosed with bicytopenia. The                   predominantly small megakaryocytes. FISH analysis was
CBC was as follows: WBC 3.3 × 109/L, Hb 7.8 g/dL, MCV                     consistent with del(5). No other cytogenetic abnormalities
111 fL, platelets 159 × 109/L. The peripheral blood smear                 were detected. Molecular studies revealed a SF3B1 muta-
showed anisocytosis of platelets and some hypolobated neu-                tion. The diagnosis was myelodysplastic syndrome with
trophils but no blasts. The bone marrow aspirate smear                    del(5q), concomitant SF3B1 mutation, and ring sideroblasts.
showed mild dysplasia of neutrophil precursors, hypolo-                   Cases with these findings are included in the category MDS
bated megakaryocytes, and a megaloblastoid erythropoiesis                 with del(5q).
a b
MGG MGG
c d
PB PB
Fig. 12.11 Myelodysplastic syndrome with del(5q), concomitant             stain). (e, f) Hypercellular bone marrow core biopsy with increased
SF3B1 mutation, and ring sideroblasts. (a, b) Bone marrow aspirate        macroblastic erythroid precursors and numerous small megakaryocytes
smear showing mild dysplasia of the granulocytic lineage in a and small   (Giemsa). (g, h) Presence of megakaryocytes with hypolobated and
hypolobated megakaryocytes in b (MGG). (c, d) Markedly increased          non-lobated megakaryocytes demonstrated by CD61 immunostaining
bone marrow iron and presence of ring sideroblasts (Prussian blue
12   Myelodysplastic Syndromes (MDS)                    369
e f
Giemsa Giemsa
g h
CD61 CD61
Case History 10. Hypoplastic Myelodysplastic                               etic precursors including 7% blasts. The core biopsy was
Syndrome with Excess Blasts 1 (hMDS-EB-1)                                  markedly hypocellular with focal presence of active hemato-
(Fig. 12.12)                                                               poiesis with reduced granulopoiesis, some erythroid islands,
A 67-year-old male patient was diagnosed with pancytope-                   and small dysplastic megakaryocytes. In addition, scattered
nia. The CBC was as follows: WBC 2.1 × 109/L, Hb 6.1 g/                    CD34+ blasts were seen representing <10% of nucleated
dL, MCV 102 fL, platelets 45 × 109/L. The peripheral blood                 cells. Cytogenetic analysis revealed del5(q) and monosomy
showed 34% dysplastic neutrophils, 1% eosinophil, 4%                       7. In addition, a SRSF2 mutation was detected. The diagnosis
monocytes, and 61% lymphocytes. The bone marrow aspi-                      was hypoplastic myelodysplastic syndrome with excess
rate smear contained abundant fat cells and rare hematopoi-                blasts 1 (MDS-EB-1).
a b
                                                                MG
                                                                M
                                                                MGG
                                                                  GG                                                                    MGG
                                                                                                                                        MG
                                                                                                                                         GG
  c                                                                        d
MPO MPO
Fig. 12.12 Hypoplastic myelodysplastic syndrome with excess                (myeloperoxidase). (e, f) Hypocellular bone marrow core biopsy show-
blasts 1 (MDS-EB-1). (a, b) Nuclear hypolobulation and decreased           ing focal hematopoiesis with small dysplastic megakaryocytes
cytoplasmic granules of neutrophils in the peripheral blood. Presence of   (Giemsa). (g) Rare cells of granulopoiesis are naphthol-ASD-
some pseudo-Pelger forms. (c, d) Bone marrow aspirate smear showing        chloroacetate-esterase positive while small erythropoietic islands are
abundant fat cells and rare predominantly immature myeloid cells           negative. (h) Scattered blasts are visualized by CD34 immunostaining
12    Myelodysplastic Syndromes (MDS)                  371
e f
H&E H&E
g h
NASDCL CD34
Case History 11. Hypoplastic Myelodysplastic                                The bone marrow core biopsy was hypocellular with focal
Syndrome with Excess Blasts 2 and Erythroid                                 “hot spots” of predominantly composed of erythroid cells.
Predominance (hMDS-EB-2) (Fig. 12.13)                                       Erythropoiesis represented 65% of bone marrow cells. The
A 69-year-old male patient was diagnosed with bicytopenia.                  percentage of CD34+ blasts was 14% calculated on all nucle-
The CBC was as follows: WBC 1.7 × 109/L, Hb 9.3 g/dL,                       ated cells. The karyotype was normal 46,XY. Molecular
MCV 98 fL, platelets 52 × 109/L. The peripheral blood con-                  analysis revealed a SRSF2 mutation. The diagnosis was
tained 19% neutrophils, 4% monocytes, 1% blasts, and 77%                    hypoplastic myelodysplastic syndrome with excess blasts 2
lymphocytes. The aspirate smears could not be evaluated.                    and erythroid predominance (MDS-EB-2).
a b
                                                                  H&E
                                                                  H&                                                                      H&E
                                                                                                                                          H&
                                                                                                                                          H&E
c d
                                                                  H&E
                                                                  H&                                                                      H&E
                                                                                                                                          H&
                                                                                                                                           &E
Fig. 12.13 Hypoplastic myelodysplastic syndrome with excess                 bone marrow cells. (f) Small dysplastic hypolobated CD61+ megakary-
blasts 2 and erythroid predominance. (a–d) H&E stained sections cut         ocytes in the vicinity of a lymphoid aggregate. (g) Rare left-shifted
from the hypocellular bone marrow core biopsy showing predominance          myeloperoxidase+ myloid precursors. (h) Clusters of CD34+ blasts in
of erythroid cells and small clusters of immature precursors/blasts in d.   the central bone marrow area
(e) Increased CD71+ erythroid precursors accounting for about 65% of
12   Myelodysplastic Syndromes (MDS)                373
e f
CD71 CD61
g h
                                       MPO
                                       MP
                                       MPOO       CD34
                                                  CD
                                                  CD34
Case History 12. Overlap Between Aplastic Anemia and                    composed of immature erythroid precursors. Myeloid cells
Hypoplastic MDS (Fig. 12.14)                                            and megakaryocytes were rare. Focal lymphoid infiltrates
A 28-year-old woman was admitted to the hospital with a                 and an excessive storage of hemosiderin were present. No
history of pancytopenia diagnosed several years previously.             excess blasts was observed. The karyotype was normal.
She had received multiple red blood cell transfusions. The              Molecular analysis revealed a somatic DNMT3A mutation
CBC was as follows: WBC 1.3 × 109/L, Hb 5.6 g/dL, MCV                   (Exon 23: c.2645 G->A, p.R882H). No germline mutations
86.4 fL, platelets 24 × 109/L. The differential count showed            were detected. The differential diagnosis included very
2% band forms, 38% neutrophils, 6% monocytes, and 52%                   severe aplastic anemia and hypoplastic MDS. Both condi-
lymphocytes. Some dysplastic neutrophils including pseudo-             tions may harbor a DNMT3A mutation. The final diagnosis
Pelger forms were observed. The bone marrow aspirate                    favored very severe aplastic anemia associated with an
resulted in a dry tap. The bone marrow core biopsy was                  increased risk of developing a MDS as a consequence of the
markedly hypocellular with several hot spots predominantly              DNMT3A mutation.
a b
                                                                                                                                   Gie
                                                                                                                                     em
                                                                                                                                      mssa
                                                                                                                                   Giemsaa
                                                          Gie
                                                          Giemmsa
                                                               sa
c d
                                                          Giem
                                                          Giemsa
                                                            em sa                                                                  Giemsa
                                                                                                                                   Gi
                                                                                                                                   G iem
                                                                                                                                      emsa
                                                                                                                                        s
Fig. 12.14 Overlap between aplastic anemia and hypoplastic MDS.         karyocytes by CD61 immunolabeling. (g) Rare predominantly immature
(a–d) Markedly hypocellular bone marrow core biopsy showing some        cells of the granulocytic series (naphthol-ASD-chloroacetate-esterase).
hot spots composed of immature erythroid precursors (Giemsa). (e)       (h) Rare CD34+ precursor/blast cells
CD71 immunoreactivity of erythroid cells. (f) Detection of rare mega-
12   Myelodysplastic Syndromes (MDS)                   375
e f
CD71 CD61
g h
                                       NASDCL
                                       NAS
                                       NA SD
                                           DCL
                                            C        CD34
                                                     CD
                                                     CD34
                                                      D34
                                                       34
Case History 13: Hypoplastic MDS with Multilineage                       myeloid cells were observed. Megakaryopoiesis was highly
Dysplasia and Fibrosis Grade 2 (Fig. 12.15)                              dysplastic with the presence of small hypolobated forms
A 62-year-old woman was diagnosed with severe pancyto-                    and some megakaryoblasts. CD34 immunostain revealed
penia. The CBC was as follows: WBC 0.8 × 109/L, Hb 5.1 g/                 rare blast cells accounting for about 4% of nucleated cells.
dL, MCV 101 fL, platelets 18 × 109/L. The differential                    Furthermore, interstitial CD3+ lymphocytes were slightly
count showed 12% neutrophils, 1% eosinophils, 3% mono-                    increased. The karyotype was complex and included a sub-
cytes, and 84% lymphocytes. A bone marrow core biopsy                     clone carrying a del(5q). NGS provided evidence of a TP53
was markedly hypocellular and showed an edematous mar-                    mutation. The diagnosis was hypoplastic myelodysplastic
row with an increased reticulin fiber network consistent                  syndrome with multilineage dysplasia and fibrosis grade 2
with reticulin fibrosis grade 2. Hot spots with dysplastic ery-           (hMDS-MLD-F) associated with an unfavorable prognostic
throid colonies and scattered predominantly immature                      score.
a b
H&E H&E
c d
H&E CD61
Fig. 12.15 Hypoplastic MDS with multilineage dysplasia and fibrosis      CD61 immunostain. (e) Expression of CD71 by atypically localized
grade 2. (a–c) H&E stained section of the bone marrow core biopsy        erythroid colonies. (f) Rare immature myeloid cells are positive for
showing hypocellular edematous marrow spaced containing a strikingly     myeloperoxidase. (g) CD34 immunolabeling of rare myeloid blasts and
diminished hematopoiesis. Note the presence of a focally enlarged dys-   some micromegakaryocytes. (h) Gomori stain consistent with fibrosis
plastic erythroid colony and of small megakaryocytes with hypolobated    grade 2
nuclei in c. (d) Presence of micromegakaryocytes is highlighted by
12   Myelodysplastic Syndromes (MDS)                  377
e f
CD71 MPO
g h
CD34 Gomori
Case History 14: Myelodysplastic Syndrome Excess Blasts 2               showed markedly dysplastic megakaryocytes with com-
(MDS-EB-2) Progressing to Early AML with                                pletely separated nuclei, macroblastic erythroid precursors,
Myelodysplasia-Related Changes (AML MRC) (Fig. 12.16)                   rare myeloid precursors, and 22% blasts highlighted by
A 84-year-old man was diagnosed with pancytopenia. The                  CD34 immunolabeling. Cytologic examination of aspirate
CBC was as follows: WBC 1.2 × 109/L, Hb 5.2 g/dL, MCV                   smears revealed blasts with coarse myeloperoxidase+ gran-
98 fL, platelets 65 × 109/L. The peripheral blood smears                ules and rare small Auer rods. The karyotype was complex.
showed 43% hypogranulated neutrophils, 11% monocytes,                   The diagnosis was myelodysplastic syndrome excess blasts 2
1% eosinophils, 1% basophils, and 44% lymphocytes. Bone                 transforming into early AML with myelodysplasia-related
marrow aspirate smears and trephine bone marrow biopsy                  changes (AML-MRC).
a b
                                                                                                                                       H&E
                                                              H&E
c d
                                                              H
                                                              H&
                                                              H&E
                                                               &E                                                                       H&E
Fig. 12.16 Myelodysplastic syndrome excess blasts 2 (MDS-EB-2)          Enlarged macroblastic erythroid colony (hemoglobin). (f) Atypically
progressing to early AML with myelodysplasia-related changes (AML-     localized clusters of CD34+ blast cells in the vicinity to CD34+ vascu-
MRC). (a–d) Age-related slightly hypercellular bone marrow showing      lar endothelium. (g, h) Bone marrow aspirate smears showing blasts
enlarged macroblastic erythroid colonies close to atrophic trabecular   containing myeloperoxidase+ granules and rare small Auer rods (MPO)
bone and small megakaryocytes with bilobated nuclei (H&E). (e)
12   Myelodysplastic Syndromes (MDS)                379
e f
                                        HB
                                        HB        CD34
g h
                                       M
                                       MPPO
                                       MPOO       MPO
                                                  MPO
References                                                                 16. Valent P, Orazi A, Steensma DP, Ebert BL, Haase D, Malcovati
                                                                               L, van de Loosdrecht AA, Haferlach T, Westers TM, Wells DA,
                                                                               Giagounidis A, Loken M, Orfao A, Lübbert M, Ganser A, Hofmann
 1. Arber DA, Orazi A, Hasserjian R, Thiele J, Borowitz MJ, Le Beau            WK, Ogata K, Schanz J, Béné MC, Hoermann G, Sperr WR, Sotlar
    MM, Bloomfield CD, Cazzola M, Vardiman JW. The 2016 revi-                  K, Bettelheim P, Stauder R, Pfeilstöcker M, Horny HP, Germing U,
    sion to the World Health Organization classification of myeloid            Greenberg P, Bennett JM. Proposed minimal diagnostic criteria for
    neoplasms and acute leukemia. Blood. 2016;127(20):2391–405.                myelodysplastic syndromes (MDS) and potential pre-MDS condi-
    https://doi.org/10.1182/blood-2016-03-643544. Epub 2016 Apr 11.            tions. Oncotarget. 2017;8(43):73483–500.
    Review.
                                                                           17. Baumann I, Führer M, Behrendt S, Campr V, Csomor J, Furlan I, de
 2. Hasserdjian RP, Orazi A, Brunning RD, Germing U, Le Beau MM,
                                                                               Haas V, Kerndrup G, Leguit RJ, De Paepe P, Noellke P, Niemeyer C,
    Porwit A, Baumann I, Hellström-Lindberg E, List AF, Cazzola M,
                                                                               Schwarz S. Morphological differentiation of severe aplastic anae-
    Foucar K. Myelodysplstic syndromes. In: Swerdlow SH, Campo
                                                                               mia from hypocellular refractory cytopenia of childhood: repro-
    E, Harris NL, Jaffe ES, Pileri SA, Stein H, Thiele J, editors. WHO
                                                                               ducibility of histopathological diagnostic criteria. Histopathology.
    classification of tumours of haematopoietic and lymphoid tissues.
                                                                               2012;61(1):10–7.
    4th ed. Lyon: International Agency for Research on Cancer; 2017.
                                                                           18. Kvasnicka HM, Beham-Schmid C, Bob R, Dirnhofer S, Hussein
    p. 98–106.
                                                                               K, Kreipe H, Kremer M, Schmitt-Graeff A, Schwarz S, Thiele J,
 3. Zini G. Diagnostics and prognostication of myelodysplas-
                                                                               Werner M, Stein H. Problems and pitfalls in grading of bone mar-
    tic syndromes. Ann Lab Med. 2017;37(6):465–74. https://doi.
                                                                               row fibrosis, collagen deposition and osteosclerosis—a consensus-
    org/10.3343/alm.2017.37.6.465. Review.
                                                                               based study. Histopathology. 2016;68(6):905–15. https://doi.
 4. Weinberg OK, Hasserdjian RP. The current approach to the diagnosis
                                                                               org/10.1111/his.12871. Epub 2015 Nov 20.
    of myelodysplastic syndromes. Semin Hematol. 2019;56(1):15–21.
                                                                           19. Della Porta MG, Malcovati L, Boveri E, Travaglino E, Pietra D,
 5. Arber DA, Brunning RD, Le Beau MM, Falini B, Vardiman JW,
                                                                               Pascutto C, et al. Clinical relevance of bone marrow fibrosis and
    Porwit A, Thiele J, Foucar K, Döhner H, Bloomfield CD. Acute
                                                                               CD34-positive cell clusters in primary myelodysplastic syndromes.
    leukemia with recurrent cytogenetic abnormalities. In: Swerdlow
                                                                               J Clin Oncol. 2009;27(5):754–62.
    SH, Campo E, Harris NL, Jaffe ES, Pileri SA, Stein H, Thiele J,
    editors. WHO classification of tumours of haematopoietic and lym-      20. Angelucci E, Cianciulli P, Finelli C, Mecucci C, Voso MT, Tura
    phoid tissues. 4th ed. Lyon: International Agency for Research on          S. Unraveling the mechanisms behind iron overload and ineffec-
    Cancer; 2017. p. 130–49.                                                   tive hematopoiesis in myelodysplastic syndromes. Leuk Res.
 6. Bennett JM, Catovsky D, Daniel MT, et al. Proposed revised cri-            2017;62:108–15.
    teria for the classification of acute myeloid leukemia. A report of    21. Tao J, Li L, Wang Y, Fu R, Wang H, Shao Z. Increased TIM3+CD8+T
    the French-American-British cooperative group. Ann Intern Med.             cells in myelodysplastic syndrome patients displayed less perforin
    1985;103:626–9.                                                            and granzyme B secretion and higher CD95 expression. Leuk Res.
 7. Della Porta MG, Travaglino E, Boveri E, Ponzoni M, Malcovati               2016;51:49–55.
    L, Papaemmanuil E, Rigolin GM, Pascutto C, Croci G, Gianelli           22. Braulke F, Platzbecker U, Müller-Thomas C, Götze K, Germing
    U, Milani R, Ambaglio I, Elena C, Ubezio M, Da Via’ MC, Bono               U, Brümmendorf TH, Nolte F, Hofmann WK, Giagounidis AA,
    E, Pietra D, Quaglia F, Bastia R, Ferretti V, Cuneo A, Morra E,            Lübbert M, Greenberg PL, Bennett JM, Solé F, Mallo M, Slovak
    Campbell PJ, Orazi A, Invernizzi R, Cazzola M, Rete Ematologica            ML, Ohyashiki K, Le Beau MM, Tüchler H, Pfeilstöcker M,
    Lombarda (REL) Clinical Network. Minimal morphological cri-                Nösslinger T, Hildebrandt B, Shirneshan K, Aul C, Stauder R,
    teria for defining bone marrow dysplasia: a basis for clinical             Sperr WR, Valent P, Fonatsch C, Trümper L, Haase D, Schanz
    implementation of WHO classification of myelodysplastic syn-               J. Validation of cytogenetic risk groups according to international
    dromes. Leukemia. 2015;29(1):66–75. https://doi.org/10.1038/               prognostic scoring systems by peripheral blood CD34+FISH:
    leu.2014.161.                                                              results from a German diagnostic study in comparison with an
 8. DeZern AE. Treatments targeting MDS genetics: a fool's errand?             international control group. Haematologica. 2015;100(2):205–
    Hematol Am Soc Hematol Educ Program. 2018;2018(1):277–85.                  13. https://doi.org/10.3324/haematol.2014.110452.
 9. Hasle H. Myelodysplastic and myeloproliferative disorders of child-    23. Greenberg PL, Tuechler H, Schanz J, Sanz G, Garcia-Manero G,
    hood. Hematol Am Soc Hematol Educ Program. 2016;2016(1):598–               Solé F, Bennett JM, Bowen D, Fenaux P, Dreyfus F, Kantarjian
    604. Review.                                                               H, Kuendgen A, Levis A, Malcovati L, Cazzola M, Cermak J,
10. Locatelli F, Strahm B. How I treat myelodysplastic syndromes of            Fonatsch C, Le Beau MM, Slovak ML, Krieger O, Luebbert M,
    childhood. Blood. 2018;131(13):1406–14. Epub 2018 Feb 8.                   Maciejewski J, Magalhaes SM, Miyazaki Y, Pfeilstöcker M,
11. Galaverna F, Ruggeri A, Locatelli F. Myelodysplastic syndromes in          Sekeres M, Sperr WR, Stauder R, Tauro S, Valent P, Vallespi T,
    children. Curr Opin Oncol. 2018;30(6):402–8.                               van de Loosdrecht AA, Germing U, Haase D. Revised international
12. Nazha A. The MDS genomics-prognosis symbiosis. Hematol Am                  prognostic scoring system for myelodysplastic syndromes. Blood.
    Soc Hematol Educ Program. 2018;2018(1):270–6. https://doi.                 2012;120(12):2454–65.
    org/10.1182/asheducation-2018.1.270. Review.                           24. Papaemmanuil E, Gerstung M, Malcovati L, Tauro S, Gundem G,
13. Della Porta MG, Picone C, Tenore A, Yokose N, Malcovati                    Van Loo P, Yoon CJ, Ellis P, Wedge DC, Pellagatti A, Shlien A,
    L, Cazzola M, Ogata K. Prognostic significance of repro-                   Groves MJ, Forbes SA, Raine K, Hinton J, Mudie LJ, McLaren
    ducible immunophenotypic markers of marrow dysplasia.                      S, Hardy C, Latimer C, Della Porta MG, O'Meara S, Ambaglio
    Haematologica.        2014;99(1):e8–10.     https://doi.org/10.3324/       I, Galli A, Butler AP, Walldin G, Teague JW, Quek L, Sternberg
    haematol.2013.097188.                                                      A, Gambacorti-Passerini C, Cross NC, Green AR, Boultwood J,
14. Schmitt-Graeff A. Myelodysplastic syndromes an acute myeloid               Vyas P, Hellstrom-Lindberg E, Bowen D, Cazzola M, Stratton MR,
    leukemia: morphologic and phenotypic aspects. In: Lübbert M,               Campbell PJ, Chronic Myeloid Disorders Working Group of the
    Gore SD, editors. Myelodysplastic syndromes an acute myeloid               International Cancer Genome Consortium. Clinical and biological
    leukemia: a biological and therapeutic continuum 2015. London,             implications of driver mutations in myelodysplastic syndromes.
    Boston: UNI-MED Verlag AG Bremen; 2014. p. 46–73.                          Blood. 2013;122(22):3616–27.
15. Pronk E, Raaijmakers MHGP. The mesenchymal niche in                    25. da Silva-Coelho P, Kroeze LI, Yoshida K, Koorenhof-Scheele TN,
    MDS. Blood. 2019;133(10):1031–8. pii: blood-2018-10-844639.                Knops R, van de Locht LT, de Graaf AO, Massop M, Sandmann
12    Myelodysplastic Syndromes (MDS)                                                                                                      381
      S, Dugas M, Stevens-Kroef MJ, Cermak J, Shiraishi Y, Chiba K,           dromes. Blood. 2015;126(1):9–16. https://doi.org/10.1182/
      Tanaka H, Miyano S, de Witte T, Blijlevens NMA, Muus P, Huls            blood-2015-03-631747.
      G, van der Reijden BA, Ogawa S, Jansen JH. Clonal evolution in    30.   Malcovati L, Karimi M, Papaemmanuil E, Ambaglio I, Jädersten M,
      myelodysplastic syndromes. Nat Commun. 2017;8:15099. https://           Jansson M, Elena C, Gallì A, Walldin G, Della Porta MG, Raaschou-
      doi.org/10.1038/ncomms15099.                                            Jensen K, Travaglino E, Kallenbach K, Pietra D, Ljungström V, Conte
26.   Ogawa S. Genetics of MDS. Blood. 2019;133(10):1049–59.                  S, Boveri E, Invernizzi R, Rosenquist R, Campbell PJ, Cazzola M,
      https://doi.org/10.1182/blood-2018-10-844621. pii: blood-2018-         Hellström Lindberg EB. SF3B1 mutation identifies a distinct sub-
      10-844621. [Epub ahead of print].                                      set of myelodysplastic syndrome with ring sideroblasts. Blood.
27.   Malcovati L, Cazzola M. The shadowlands of MDS: idiopathic              2015;126(2):233–41. https://doi.org/10.1182/blood-2015-03-633537.
      cytopenias of undetermined significance (ICUS) and clonal hema-   31.   Chang CK, Zhao YS, Xu F, Guo J, Zhang Z, He Q, Wu D, Wu LY,
      topoiesis of indeterminate potential (CHIP). Hematol Am Soc             Su JY, Song LX, Xiao C, Li X. TP53 mutations predict decitabine-
      Hematol Educ Program. 2015;2015:299–307.                                induced complete responses in patients with myelodysplastic syn-
28.   Tanaka TN, Bejar R. MDS overlap disorders and diagnostic bound-         dromes. Br J Haematol. 2017;176(4):600–8.
      aries. Blood. 2019;133(10):1086–95. https://doi.org/10.1182/      32.   Bejar R. What biologic factors predict for transformation to AML?
      blood-2018-10-844670. pii: blood-2018-10-844670.                        Best Pract Res Clin Haematol. 2018;31(4):341–5.
29.   Steensma DP, Bejar R, Jaiswal S, Lindsley RC, Sekeres MA,         33.   Platzbecker U. Treatment of MDS. Blood. 2019;133(10):1096–
      Hasserjian RP, Ebert BL. Clonal hematopoiesis of indetermi-             107. https://doi.org/10.1182/blood-2018-10-844696. pii: blood-
      nate potential and its distinction from myelodysplastic syn-            2018-10-844696. [Epub ahead of print].
                                 Acute Leukemia of Myeloid, Lymphoid,
                                 and Ambiguous Lineage and Related                                                            13
                                 Malignancies
Table 13.1 Important clinical and laboratory features and diagnostic   Table 13.2 AML and related disorders according to the Revised 2016
procedures in patients with suspected AL                               WHO Classification (adopted from Arber et al. [2])
Important aspects          Features, techniques                        AML with recurrent genetic abnormalities
Clinical presentation      Symptoms: weakness, bleeding,                AML with t(8;21)(q22;q22.1); RUNX1-RUNX1T1
                           infections, skin or mucosal infiltrates,     AML with inv(16)(p13.1q22) or t(16;16)(p13.1;q22);
                           hepatosplenomegaly, lymphadenopathy,          CBFB-MYH11
                           age-related performance status               Acute promyelocytic leukemia with PML-RARA
Patient history            Medical history: antecedent cytopenias,      AML with t(9;11)(p21.3;q23.3); MLLT3-KMT2A
                           recurrent infections, prior cytotoxic        AML with t(6;9)(p23;q34.1); DEK-NUP214
                           therapy, exposure to toxins, irradiation,    AML with inv(3)(q21.3;q26.2) or t(3;3)(q21.3;q26.2);
                           demographics                                  GATA2,MECOM(EVI1)
Family history             Hematologic disorders,                       AML (megakaryoblastic) with t(1;22)(p13.3;q13.1);
                           immunodeficiency states                       RBM15-MKL1
Peripheral blood           Complete blood count including               Provisional entity: AML with BCR-ABL1
                           differential count, cytologic evaluation
                                                                       AML with gene mutations
                           of ≥200 white blood cells on blood
                                                                        AML with mutated NPM1
                           smears, immunocytochemistry,
                           immunophenotyping, molecular                 AML with biallelic mutations of CEBPA
                           analyses (see below)                         Provisional entity: AML with mutated RUNX1
Bone marrow                Aspirate: microscopic evaluation of         AML with myelodysplasia-related changes
                           ≥500 nucleated cells with special regard    Therapy-related myeloid neoplasms
                           to blast cells on smears; trephine bone     AML, NOS
                           marrow biopsy: overall cellularity,             AML with minimal differentiation
                           hematopoietic compartment, atypical             AML without maturation
                           cells, stroma including fibrosis                AML with maturation
                           Additional techniques (see below)               Acute myelomonocytic leukemia
Additional laboratory data Lactate dehydrogenase (LDH),                    Acute monoblastic/monocytic leukemia
                           coagulation tests, liver function tests        Pure erythroid leukemia
Immunophenotype            Cell surface and cytoplasmic markers            Acute megakaryoblastic leukemia
                           by flow cytometry (liquid samples),
                                                                           Acute basophilic leukemia
                           immunocytochemistry and/or
                           immunohistochemistry (smears, biopsy            Acute panmyelosis with myelofibrosis
                           sections) using adequate antibody           Myeloid sarcoma
                           panels                                      Myeloid proliferations related to Down syndrome
Gross chromosomal          Karyotyping (metaphase chromosomes              Transient abnormal myelopoiesis
alterations                from viable bone marrow or blood                Myeloid leukemia associated with Down syndrome
                           cells), fluorescence or chromogen in situ   Blastic plasmacytoid dendritic cell neoplasm
                           hybridization (interphase nuclei of bone    Acute leukemias of ambiguous lineage
                           marrow aspirate or blood smears,             Acute undifferentiated leukemia
                           trephine bone marrow biopsy sections)        MPAL with t(9;22)(q34.1;q11.2); BCR-ABL1
Recurrent genetic          Sequencing technologies including            MPAL with t(v;11q23.3); KMT2A rearranged
mutations, epigenetics     Sanger sequencing, next-generation           MPAL, B/myeloid, NOS
                           targeted or whole genome sequencing,
                                                                        MPAL, T/myeloid, NOS
                           epigenomics (liquid samples or
                           formalin-fixed paraffin-embedded            In addition, various myeloid neoplasms with germline predisposition
                           material)                                   with or without a preexisting disorder or organ dysfunction are listed in
                                                                       the WHO classification [2, 16]
sarcoma [15], and myeloid proliferations associated with                  In AML patients, the genetic and mutational profiles that
Down syndrome [16].                                                    are involved in disease pathogenesis are important for dis-
   At initial presentation, AML is generally a bone                    ease classification and prognostic stratification [2, 17–19].
marrow-based neoplasm associated with the expansion of                The European Leukemia Net (ELN) has defined four catego-
≥20% blast cells but may also involve extramedullary                   ries of AML risk based on six fusion genes, three genes with
sites. Exceptions are cases with low blast counts present-             point mutations and cytogenetic abnormalities [20]. The
ing with AML-type recurrent cytogenetic abnormalities                  ELN recommendations have actually been refined [6]. In the
that are straightforwardly classified as AML (Table 13.2).             future, individual patient outcome may be predicted from
AML with t(8;21)(q22;q22.1); RUNX1-RUNX1T1, t(15;17)                   genomic and clinical variables that may be important tools
(q22;q11-12); PML-RARA, and AML with inv(16)                           for precision oncology [21]. However, the phenotype of the
(p13.1q22) or t(16;16)(p13.1;q22); CBFB-MYH11 are                      predominant blast cell population still remains pivotal for
considered as AML regardless of the percentage of blasts               AML cases lacking specific molecular and/or cytogenetic
in the bone marrow.                                                    features. They are included in the “NOS” category of AML.
13   Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies                                                   385
Table 13.4 Routine immunohistochemical stains on BM biopsies in        Table 13.5 Routine genetic testing by FISH in AML
AML
                                                                       FISH probes to detect chromosomal translocations, rearrangements,
Antigen                Cell type                     Comments          and deletions
Myeloperoxidase        Myeloblasts±,                                    Translocation t(15;17)/PML-RARA
(MPO)                  promyelocytes+, mature                           Translocation t(8;21)/RUNX1-RUNX1T1(AML1-ETO)
                       neutrophils+, monocytes/                         Translocation t(6;9)/DEK-NUP214
                       promonocytes(±),monoblasts−                      CBFβ-Rearrangement/inv(16), t(16;16)
CD34                   Myeloid precursors+,          Low or absent      KMT2a/MLL (11q23)-Rearrangement
                       myeloblasts+, endothelial     expression in      MECOM (3q26)-Rearrangement
                       cells+                        APL
                                                                        Deletion 5q31/5q33 (EGR1)/RPS14
CD117                  Mast cells+,
                                                                        Monosomy 7/Deletion 7q22, 7q31,7q36
                       myeloblasts±, monoblasts
                       (−/+)                                            Deletion 17p13 (TP53)
CD33, CD13, CD15       Myeloblasts+,                 CD33: strong       Deletion 20q12
                       promyelocytes+                expression in     Analysis of 100 interphase nuclei is mandatory
                                                     AML with
                                                     mutated NPM1
HLA-DR                 Myeloblasts+,                 Low or absent     can be applied to aspirate smears as well as to formalin-fixed
                       promyelocytes+,               expression in     paraffin embedded (FFPE) tissue sections. According to the
                       monoblasts+,                  APL               2017 ELN recommendations, FISH should be performed to
                       promonocytes+
                                                                       detect gene rearrangements, such as RUNX1-RUNX1T1,
CD14, CD64, CD68       Monoblasts+,
CD11c, CD11c,          promonocytes+                                   CBFB-MYH11, KMT2A (MLL), and MECOM (EVI1) gene
CD36, lysozyme                                                         fusions, or loss of chromosome 5q, 7q, or 17p material if con-
CD71, glycophorin CErythroblasts                                       ventionally cytogenetic testing is not successful [6].
                   (pronormoblasts)                                       Translocations/inversions are detected in ~50% and 30%
CD41, CD61, CD42b, Megakaryoblasts                                     of AML arising in children and younger adults but are less
CD31
CD7                T-cells                           May aberrantly
                                                                       frequent in older patients [18]. In about 45% adult AML
                                                     be expressed in   cases, no cytogenetic abnormalities can be detected. These
                                                     AML               cases are referred to as the category cytogenetically normal
                                                                       (CN) AML. Rare children with normal karyotype by conven-
                                                                       tional cytogenetic evaluation harbor socalled cryptic rear-
or histological specimens as well as by multiparameter flow            rangements. In addition, some cases of PML-RARA+APL
cytometry of fluid samples is mandatory for the evaluation of          lack the classic t(15;17).
blast cells and the delineation of cell lineages ([7], Table 13.4).       In the actual WHO classification, eight balanced transloca-
Blast/precursor cells are highlighted by the expression of             tions and inversions and their variants are listed [11].
markers such as CD34, CD117, CD33, HLA-DR. Note that                   Reciprocal translocations result in chimeric fusion genes
blasts of monocytic lineage often lack CD34 expression.                involving for example RARA, RUNX1, the CBFβ subunits of
Myeloid lineage antigens are cytoplasmic MPO, CD13,                    the core binding factor complex, epigenetic regulators such as
CD33, CD15, and CD65. Monocytic lineage markers are                    KMT2A formerly designated as MLL, or components of the
CD14, CD36, CD64, frequently also CD4 and CD56.                        nuclear pore complex and other genes [18]. The detection of
   Megakaryocytic markers are CD41, CD42b and CD61.                    certain recurrent genetic abnormalities in blood and/or mar-
Erythroid markers are CD235a (glycophorin A), CD36, CD71,              row samples allows a diagnosis of AML without regard to the
and often weak CD117 [6, 32]. Flow cytometry is also helpful           blast cell count. The AML-defining recurrent cytogenetic
for the evaluation of minimal residual disease in AML [33].            abnormalities actually include t(8;21)(q22;q22.1); RUNX1-
                                                                       RUNX1T1; inv(16)(p13.1q22) or t(16;16)(p13.1;q22); CBFB-
                                                                       MYH11; t(15; 17)(q24.1;q21.2); PML-RARA or variant
13.2.5 Genetics                                                       translocations and t(1;22)(p13.3;q13.3); RBM15-MKL1.
                                                                          The detection of nine balanced rearrangements and multiple
In 1973, cytogenetic studies performed by Janet Rowley                 unbalanced abnormalities allows the classification of a myeloid
revealed that subgroups of AML are associated with the bal-            neoplasm with ≥20% blood or marrow blasts as “AML with
anced chromosomal rearrangements t[8;21] or t[15;17] first             myelodysplasia-related changes (AML-MRC)” [6].
indicating that AML is a genetic disease [34–36].                         Based on cytogenetic findings, a risk stratification has been
   Karyotype analyses can be performed either by conven-               established including favorable groups [t(8;21)(q22;q22.1);
tional cytogenetic methods applied to metaphase chromosomes            inv(16)(p13.1q22) or t(16;16)(p13.1;q22); t(15; 17)
or by molecular cytogenetic methods performed on non-divid-            (q24.1;q21.2)], intermediate groups [normal karyotype, all
ing cells (Table 13.5). A widely used method is the fluorescence       prognostically not favorable or adverse types], and adverse
in situ hybridization (FISH) technique using a large panel of          groups [numerous karyotypes including inv(3)/t(3;3); mono-
commercially available fluorescently labeled DNA probes that           somal karyotype (loss of ≥2 unrelated abnormalities); complex
13   Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies                                                387
karyotype (≥3 unrelated abnormalities) and many other cyto-          Table 13.6 Some important cytogenetic/molecular genetic alterations
genetic aberrations]. The cytogenetic risk stratification is vari-   routinely tested in AML
able according to the age of patients.                               Genetic alteration Cytogenetics              Relevance for outcome
   Multiple techniques including Sanger sequencing of can-           PML/RARA           t(15; 17); Cryptic and    Early complications due
                                                                                        variant translocations    to coagulopathy, good
didate genes and high-throughput sequencing techniques                                  may occur                 response to all-trans
have contributed to the understanding of the complex                                                              retinoic acid and arsenic
genomic alterations in AML with normal cytogenetics and                                                           trioxide
those with chromosomal losses or gains that were previously          RUNX1/             t(8;21)                   Favorable, but adverse
                                                                     RUNX1T1                                      effect of concomitant
poorly understood [18].
                                                                                                                  KIT mutation
   A model of AML leukemogenesis has proposed two                    CBFB/MYH11         inv(16) or t(16;16)       Favorable
classes of cooperating mutations conferring a proliferative          MECOM (EVI1)       inv3, t(3;3) but also     Unfavorable
advantage (class I mutations) and inducing a block in                overexpression     with various normal
myeloid differentiation (class II mutations) has been pro-                              and abnormal
                                                                                        karyotypes
posed [37]. Class I comprises mutations leading to activation
                                                                     BCR-ABL1           t(9;22)                   Rare in de novo AML,
of the receptor tyrosine kinase FLT3 or the RAS signaling                                                         poor response to
pathway and includes also JAK2 and SHP2. Class II com-                                                            tyrosine kinase inhibitor
prises mutations in CEBPA, MLL, NPM1, and gene fusions                                                            (TKI) therapy
resulting from t(8;21), inv(16)/t(16;16), or t(15;17).               KIT                t(8;21)                   Unfavorable in patients
                                                                                        inv(16)/t(16;16)          with concomitant t(8;21)
   In addition to somatically acquired driver mutations, sub-        FLT3-ITD           Normal karyotype          Unfavorable, adverse
clones with differing mutational composition often develop                                                        outcome concomitant
during disease evolution. Whole-genome sequencing studies                                                         with NPM1
have provided evidence that AML is not a stable but a com-           FLT3-TKD           Various karyotypes
plex and dynamic disease with multiple competing clones              (tyrosine kinase
                                                                     domain)
[17, 38]. Mutations in genes encoding epigenetic modifiers,          NPM1               Normal karyotype          Favorable in FLT3-ITD
such as DNMT3A, ASXL1, TET2, IDH1, and IDH2, are com-                                                             wild-type
monly acquired early and are present in the founding clone.          NPM1 and           Normal karyotype
Mutations involving NPM1 or signaling molecules (e.g.,               FLT3-ITD
FLT3, RAS) are typically secondary events that occur later           CEBPA              Various karyotypes        Biallelic mutation of
                                                                                                                  CEBPA: WHO entity
during leukemogenesis [38].                                                                                       with longer disease-free
   Genetic profiling including cytogenetic analyses and                                                           and favorable overall
sequencing of 111 genes resulted in a genomic and prognostic                                                      survival
classification of AML [17]. In the large AML cohort analyzed         RUNX1              Normal or abnormal        Unfavorable
                                                                                        karyotypes
in this study, NPM1-mutated AML was the largest class
                                                                     DNMT3A             Normal karyotype          Unfavorable
(accounting for 27% of the cohort), with 73% of patients also        IDH1/2             Predominantly normal      Unfavorable, dependent
carrying mutations in DNA methylation or hydroxymethyl-                                 karyotypes                on mutation type
ation genes (DNMT3A, IDH1, IDH2R140, and TET2). In the               ASXL1              Normal and abnormal       Unfavorable
chromatin–spliceosome group (18% of the cohort), mutations                              karyotypes
in genes regulating RNA splicing (SRSF2, SF3B1, U2AF1,               TET2               Normal and abnormal       Dependent on mutation
                                                                                        karyotypes                type and cytogenetics
and ZRSR2), chromatin remodelling (ASXL1, STAG2, BCOR,               WT1                Normal and abnormal       Unfavorable
KMT2A-PTD, EZH2, and PHF6), or transcription (RUNX1)                                    karyotypes
were found. A third subgroup harbored mutations in TP53,             TP53               Complex karyotypes,       Unfavorable
complex karyotype alterations, cytogenetically visible copy-                            abnormalities of
                                                                                        chromosomes 5, 7, or 17
number alterations (aneuploidies), or a combination. A fourth
small subgroup (about 1%) carried IDH2R172 mutations [17].
   Conventional karyotyping, FISH, and molecular testing             classification includes AML with mutated NPM1 and AML
of large cohorts of AML patients have provided important             with biallelic mutations of CEBPA as distinct entities that
insights into the pathogenesis, genetic complexity, and het-         have in the absence of FLT3-ITD, a relatively favorable
erogeneous prognosis of the different AML types [6, 39].             prognosis [37]. CEBPA mutation may be inherited through
According to the 2017 ELN recommendations, screening for             the germline, with the development of AML associated with
gene rearrangements such as PML-RARA, CBFB-MYH11,                    acquisition of additional mutations [18].
RUNX1-RUNX1T1, BCR-ABL1, and other fusion genes (if                     Taken together, genetic testing of all AML cases is man-
available) should be performed [40, 41]. Current interna-            datory for risk stratification and decision-making whether
tional guidelines recommend molecular testing for a limited          individual patients are eligible for conventional therapeutic
number of mutations including NPM1, CEBPA, RUNX1,                    regimens, targeted therapies, and/or candidates for alloge-
FLT3, TP53, and ASXL1 ([6], Table 13.6). The actual WHO              neic hematopoietic cell transplantation.
388                                      13   Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies
and agranulated cytoplasm. Promyelocytes containing bun-             immunophenotype (CD15− CD34− HLA-DR−) but are
dles of Auer rods are referred to as Fagot cells.                    negative for CD2.
   On peripheral blood smears, the microgranular APL sub-        •   Atypical promyelocytes as well as the PML-RARA
type is characterized by bilobed nuclei surrounded by a baso-        fusion transcript may persist for some weeks after suc-
philic cytoplasm devoid of distinct coarse granules. These           cessful induction treatment without indicating treatment
cells are negative for naphthol-ASD-chloroacetate-esterase           failure.
stain but strongly positive for myeloperoxidase cytochemical     •   Rapid diagnosis and treatment of APL is mandatory to
stains. The bone marrow, however, contains at least some             prevent fatal hemorrhagic and/or thrombotic complica-
granulated cells and/or cells with Auer rods. In the hyperba-        tions that may result from activation of both coagulation
sophilic APL variant, dark coarse granules are observed in a         and fibrinolytic pathways by granule release from
basophilic cytoplasm.                                                promyelocytes.
   Other APL cases may show non-pathognomonic blasts on          •   APL should be suspected in cases with very low WBC
peripheral blood smears while neoplastic promyelocytes are           counts and marked thrombocytopenia [46].
present in the bone marrow.                                      •   High leukocyte counts are often present in the hypogranu-
                                                                     lar APL variant that, however, is also strongly
13.3.1.4 Caution                                                     myeloperoxidase-positive.
• APL represents a diagnostic emergency since coagulopa-         •   APL entity includes numerous morphologic, immuno-
  thy and bleeding, particularly within the brain and lungs,         phenotypic, and molecular variant forms.
  are the most common causes of early death.                     •   Differential diagnosis includes promyelocytic maturation
• Some cases of acute myeloid leukemia (AML) with                    inhibition by certain drugs, growth factor therapy, and
  nucleophosmin (NPM) mutations have an APL-like                     AML with t(8;21) with densely granulated blasts.
390                                        13   Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies
Case History 1: APL                                                    dase, CD33, and CD45 (low). CD34 and HLADR were not
A 48-year-old woman was referred for the evaluation of                 expressed.
pancytopenia and skin hemorrhage. No significant past                      FISH studies provided evidence of a translocation
medical history was reported. Fibrinogen was decreased.                t(15;17)/PML-RARA in >90% of cells and a trisomy 8 in a sub-
The CBC was as follows: Hb 9.5 g/dL, RBC 2.92 × 1012/L,                clone. Molecular tests positive for a PML-RARA fusion transcript
platelets 18 × 109/L, WBC 1.2 × 109/L. The differential                (isoform BCR1) confirmed the diagnosis of acute promyelocytic
count showed 48% atypical promyelocytes and blasts, 9%                 leukemia. The bone marrow core biopsy was infiltrated by atypi-
neutrophils, 43% lymphocytes. The bone marrow aspirate                 cal promyelocytes positive for myeloperoxidase and CD33.
contained 91% promyelocytes and blast cells with coarse                Positivity for naphthol-ASD-chloroacetate-esterase was weak in
granules and Auer rods. Some cells were packed with abun-              the neoplastic population but strongly positive in the cytoplasm
dant Auer rods (faggots) (Fig. 13.1). The neoplastic cells             of macrophages that engulfed cellular debris. Erythropoiesis and
were positive for CD117 and co-expressed myeloperoxi-                  megakaryocytes were strikingly diminished (Fig. 13.1).
a b
                                                             MG
                                                             MGG                                                                     MGG
c d
                                                             MGG                                                                     MPO
                                                                                                                                     M
                                                                                                                                     MPPO
Fig. 13.1 Acute promyelocytic leukemia. (a, b) Atypical hypergranu-    ple Auer rods. (e–h) The bone marrow core biopsy is highly hypercel-
lated promyelocyte in the peripheral blood smears (MGG). (c) Bone      lular and infiltrated by neoplastic cells with a basophilic granulated
marrow aspirate smear with heavily granulated atypical promyelocytes   cytoplasm (e, f Giemsa). (g) Strong expression of CD33 by the neoplas-
and blasts. Note the presence of Auer rods (MGG). (d) MPO reaction     tic cells but (h) negativity for CD34
demonstrating the presence of coarse cytoplasmic granules and multi-
13       Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies       391
e f
Giemsa Giemsa
g h
                                                            MPO
                                                            MP
                                                            MPO                                 CD34
                                                                                                C
                                                                                                CD
                                                                                                 D34
                                                                                                  34
a b
                                                            MG
                                                            M GG                                                                    MGG
c d
                                                             MP
                                                             M PO                                                               Giemsa
                                                                                                                                G
                                                                                                                                Giie
                                                                                                                                   em
                                                                                                                                    msa
                                                                                                                                     sa
Fig. 13.2 Acute promyelocytic leukemia (APL) with PML-RARA,            trephine bone marrow biopsy shows a dense infiltration of marrow
hypogranular variant. (a) Blood smear and (b) bone marrow aspirate     spaces by myeloid blasts (d, Giemsa) that strongly express myeloper-
(MGG) show frequently bilobed blast cells with indented nuclei and a   oxidase (e, f). The majority of blasts is negative for NASDCL (g) but
basophilic predominantly agranular or finely granulated cytoplasm      expresses CD33 (h)
(MGG) that are (c) strongly positive for myeloperoxidase. (d–g) The
13       Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies     393
e f
                                                            MPO                                MPO
     g                                                               h
                                                       NASDCL
                                                       NA
                                                       NASSD
                                                           DC
                                                            CL
                                                             L                                 CD33
                                                                                               CD
                                                                                               CD333
                                                                                                   3
13.3.2 A
        cute Myelogenous Leukemia (AML)                        [50]. Other concomitant gene mutations occur in NRAS,
       with inv(3)(q21.3;q26.2) or t(3;3)                       KRAS, KIT, GATA2, SF3B1, PTBN11, NF1, CBL, or RUNX1
       (q21.3;q26.2); GATA2; MECOM                              genes, but rarely in FLT3ITD or NPM1 [51]. By using next-
                                                                generation sequencing, mutations in genes activating RAS/
AML with inv(3)(q21.3;q26.2) or t(3;3)(q21.3;q26.2) is an       receptor tyrosine kinase pathways have been found in 98%
unfavorable subtype of AML with recurrent cytogenetic           of myeloid neoplasms with inv(3)/t(3;3). Moreover, muta-
abnormalities and can develop de novo or therapy related. In    tions in genes coding for splice factors, epigenetic modifiers,
a large cohort of AML patients, 3q abnormalities were           and transcription factors are frequent in this subtype [52].
detected in 4.4% of AML patients and associated with a
poor prognosis. The diagnosis requires 20% or more blasts,      13.3.2.2 Immunophenotype
while cases with less than 20% of blasts are categorized as     The myeloid blasts from AML with inv(3)/t(3;3) usually
MDS [11]. It has been shown that despite the difference         express CD34, HLA-DR, CD13, CD33, and MPO and may
with regard to the percentage of blasts between the             express CD56 or megakaryocyte markers such as CD41 or
inv(3)/t(3;3)-harboring MDS and AML cases, the overall          CD61. However, different expression of certain antigens were
survival showed no statistical difference. AML patients may     reported in patients with de novo AML with inv(3)(q21q26.2)
present with low or normal or even frequently with high         vs. those with t(3;3)(q21.3q26.2)-containing blasts.
platelet counts [47]. Thus, in 1979 AML and thrombocythe-
mia associated with an abnormality of chromosome 3 [inv         13.3.2.3 Morphology
(3) (q21;q26.2)] were first described, suggesting that a spe-   Peripheral blood smears contain low or high numbers of
cific structural rearrangement in human chromosomes may         blast cells that may resemble blasts from other categories
be associated with abnormal megakaryocytes and increased        with the exception of APL. A characteristic feature of many
platelets in AML [48].                                          but not all cases is the presence of large atypical platelets
                                                                and/or naked megakaryocytic nuclei.
13.3.2.1 Genetics                                                 Abundant highly dysplastic small megakaryocytes are the
The molecular basis of AML with inv(3)(q21.3q26.2) or           hallmark of most bone marrow samples. Megakaryocytes are
t(3;3)(q21.3;q26.2) was revisited showing that reposition-      often monolobated with small eccentrically localized nuclei
ing of a GATA2 enhancer element leads to overexpression         or two separated nuclei. In addition, multilineage dysplasia
of the MECOM (EVI1) gene and to haploinsufficiency of           may be observed. Bone marrow cellularity is variable and
GATA2 [49].                                                     may be decreased.
    AML with inv(3)/t(3;3)(q21.3q26.2) or t(3;3)(q21.3q26.2)
is associated with aberrant expression of the stem cell regu-   13.3.2.4 Caution
lator MECOM (formerly designated as EVI1) and simultane-        • Bone marrow samples from patients with isolated dele-
ously confers GATA2 tumor suppressor haploinsufficiency.          tion 5q are also characterized by the presence of small
Other translocations of 3q26.2 are not included in this cate-     hypolobated megakaryocytes. However, these mega-
                                                                  
gory. MECOM overexpression can be detected by real-time           karyocytes are generally larger and contain rounded
quantitative PCR and confers a very poor prognosis in both        nuclei.
AML and MDS. Cases with inv(3)/t(3;3)(q21.3q26.2) or            • Since the bone marrow of patients with inv(3)/t(3;3)
t(3;3) (q21.3q26.2) rearrangements can be diagnosed by            (q21.3q26.2) or t(3;3)(q21.3q26.2) may exhibit multilin-
fluorescence in situ hybridization (FISH), but cryptic rear-      eage dysplasia, the differential diagnosis includes AML
rangements may be missed by chromosome banding analy-             with myelodysplasia-  related changes (AML-MRC). In
sis. Deregulated expression of MECOM is frequently                these cases, the demonstration of the pathognomonic
associated with monosomy 7. The cases may also harbor             genetic changes trumps the morphologic features and
complex karyotypes, deletion 5q or 11q23 abnormalities            allows a correct classification.
13       Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies                                              395
Case History: AML with t(3;3)(q21.3q26.2) (Fig. 13.3)                    blast cells 52%. The most characteristic cytologic and
A 77-year-old woman was admitted with severe anemia                      histologic BM feature was the presence of abundant
and leukopenia while the platelet count was slightly                     small megakaryocytes containing solitary or small
increased: Hb 6.9 g/dL, platelets 380 × 109/L, WBC                       eccentrically localized nuclei or two separated nuclei.
2.1 × 109/L. Differential count: neutrophils 3%, pseudo-                Cytogenetic evaluation revealed t(3;3)(q21.3q26.2) and
Pelger cells 2%, lymphocytes 37%, eosinophils 6%,                        a monosomy 7.
a b
                                                               MGG                                                                    MGG
 c
                                                                          d
                                                              MG
                                                              M GG                                                                    MGG
Fig. 13.3 AML with t(3;3)(q21.3q26.2) blood and bone marrow              partially myeloperoxidase positive. (e–h) Trephine BM biopsy showing
smears, bone marrow core biopsy. Cytologic aspect of (a, b; MGG)         (e, Giemsa) dense infiltration by blast cells associated with eosino-
blood and (c; MGG, d, MPO) bone marrow smears. (a) Pseudo-Pelger         philia, (f) rare immature NASDCL positive cells, and (g, h) abundant
cell, (b) mononuclear blast cells with agranulated cytoplasm showing     small CD61 megakaryocytes and megakaryoblasts. The presence of
some irregular cytoplasmic protrusions. (c, d) BM aspirate with numer-   small often bilobed or non-lobed megakaryocytes is a hallmark of this
ous small mono- or bilobated megakaryocytes and (d) blasts that are      AML entity
396                     13   Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies
e f
Giemsa NASDCL
g h
                                       CD61                                                             CD61
                                                                                                        CD
                                                                                                         D61
                                                                                                           61
Case History 1 (Fig. 13.4)                                              pink granules and a perinuclear clearing (“hof”). Blast cells
A 45-year-old male patient was admitted with an outside                 expressed myeloperoxidase, CD13, HLA-DR, CD117, and
diagnosis of AML. The CBC showed marked leukocytosis                    partially CD15 and PAX5. Cytogenetic studies showed t(8;21)
(WBC 30 × 109/L), anemia (Hb 7.5 g/dL), and thrombocyto-                (q22;q22.1) RUNX1-RUNX1T1. Molecular analyses provided
penia (47 × 109/L). Differential count: 87% blasts, 2% neutro-          evidence of additional KIT D816V and ASXL2 mutations.
phils, 11% lymphocytes. The basophilic cytoplasm of                        The diagnosis was AML with t(8;21)(q22;q22.1)
peripheral blood and bone marrow blasts frequently contained            RUNX1-RUNX1T1.
a b
                                                              MGG
                                                              MG                                                                        MGG
c d
                                                              MGG
                                                              MG                                                                        MGG
                                                                                                                                        MGG
                                                                                                                                        MG
e                                                                         f
Giemsa Giemsa
Fig. 13.4 AML with t(8;21)(q22;q22.1) RUNX1-RUNX1T1. (a, MGG)           granules (d). (e–h) Bone marrow spaces were infiltrated by blasts with
Peripheral blood and (b–d, MGG) bone marrow smears showing blasts       basophilic cytoplasm (e, f, Giemsa) that were positive for (g) naphthol-
with perinuclear clearing. The cytoplasm of some blasts contains pink   ASD-chloroacetate-esterase and (h) expressed CD117
13   Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies     399
     g
                                                                  h
NASDCL CD117
Case History 2: AML with t(8;21)(q22;21q22) RUNX1-          immunohistochemistry performed on the bone marrow core
RUNX1T1 and FLT3-ITD Mutation (Fig. 13.15)                   biopsy. The neoplastic population was negative for CD14,
A 62-year-old woman presented with anemia and thrombo- CD64, CD3, CD19, CD20, CD22, and CD79a. Despite the
cytopenia, suggesting a myelodysplastic syndrome (MDS). aberrant expression of CD7 and PAX5, the criteria for
The CBC was: WBC 2.8 × 109/L, Hb 8.68 g/dL, thrombo- ambiguous lineage acute leukemia specially of mixed pheno-
cytes 140 × 109/L. The blood smears showed 15% blasts type acute leukemia (MPAL) were not fulfilled since neither
while the bone marrow aspirate smear and the core biopsy CD3 nor CD19, CD79a, or CD22 was expressed.
contained about 90% blasts. The majority of blasts expressed    Genetic studies revealed t(8;21)(q22;q22.1) RUNX1-
CD34, TdT, HLA-DR, CD13, CD33, and CD7 while small RUNX1T1 and cooperative t(8;21)(q22;q22.1) RUNX1-
subpopulations were positive for naphthol-ASD-RUNX1T1 and FLT3-ITD mutation. The diagnosis was AML
chloroacetate-esterase and myeloperoxidase. A strong aber- with t(8;21)(q22;21q22) RUNX1-RUNX1T1 and a coopera-
rant co-expression of PAX5 and CD7 was detected by tive FLT3-ITD mutation (Fig. 13.5).
a b
                                                         Gie
                                                         Giemms
                                                              sa
                                                               a                                                                Giemsa
  c                                                                    d
                                                         Giiiem
                                                         G   em
                                                             e ms
                                                                sa
                                                                 a                                                            NASDCL
                                                                                                                              N
                                                                                                                              NA
                                                                                                                               ASDC
                                                                                                                                SD
                                                                                                                                 DC
                                                                                                                                  CL
                                                                                                                                   L
Fig. 13.5 AML with t(8;21)(q22;q22.1) RUNX1-RUNX1T1 and FLT3-          Giemsa). A small subpopulation of blasts was positive for NASDCL (d)
ITD mutation. The bone marrow biopsy was infiltrated by medium-        and myeloperoxidase (e). The large majority expressed CD34 (f), TdT
sized blast cells admixed with prominent eosinophil precursors (a–c,   (g), and PAX5 (h)
13       Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies     401
e f
                                                             MPO                               CD34
     g                                                                h
TdT PAX5
13.3.4 A
        cute Myeloid Leukemia with inv(16)                   ers of blasts with high CD34 and CD117 may be expressed.
       (p13.1q22) or t(16;16)(p13.1q22);                      An aberrant CD2 expression may be detected in some cases.
       CBFB-MYH11
                                                              13.3.4.4 Genetics
Acute myeloid leukemias carrying inv(16)(p13.1q22) or In patients with this AML subtype, inv(16)(p13.1q22) is
t(16;16)(p13.1q22) chromosomal abnormalities belong to much more frequently detected than t(16;16) (p13.1q22).
the core binding factor (CBF)-AMLs. They are morpho- Both inversion (16)(p13.1q22) and translocation t(16;16)
logically related to the myelomonocytic subtype with (p13.1q22) fuse the beta subunit of core binding factor
abnormal eosinophils (AML-M4eo) of the French- (CBFB) gene located in 16q22 to the MYH11 gene located in
American-British classification [11]. The eosinophil com- 16p13, resulting in a chimeric protein product. The CBFB-
ponent also harbors the specific inv(16)(p13q22) MYH11 fusion protein blocks the differentiation process of
rearrangement and thus derive from the leukemic clone leukemic cells. CBFs are transcriptional regulators contain-
[28]. High rates of complete remission and favorable out- ing a common CBFβ (CBFB) subunit associated with one of
come in comparison to other AML subtypes can be achieved the three CBFα (CBFA) members. The MYH11 codes for a
by appropriate treatment strategies [59, 60].                 smooth muscle heavy chain. The genetic alterations may be
                                                              missed by conventional karyotyping but are detected by
13.3.4.1 Epidemiology                                        FISH and RT-PCR analyses. Additional genetic alterations
AML with inv(16)(p13.1q22) or t(16;16)(p13.1q22) is more are frequent and include gains of chromosome 22 and 8 or
frequent in children and young adults than in older patients. deletion (7q). Furthermore, mutations may be present, such
This AML subtype represents up to 10% in younger AML as KIT, NRAS, KRAS, or FLT3 genes [11, 60]. Various
patients but only up to 3% in older adult patients.           genetic alterations have an impact on the prognosis. For
                                                              example, the coexistence of BCR-ABL1 and CBFB rear-
13.3.4.2 Morphology                                          rangement is associated with poor outcome and a clinical
The characteristic morphologic feature of this AML subtype course similar to that of CML-BP [61]. Myeloid neoplasms
is the presence of blasts with monocytic and myelomono- with isolated del(16q) with the deletion of CBFB but lacking
cytic features both in the peripheral blood and the bone mar- CBFB-MYH11 rearrangement occur in older patients and are
row. In addition, the bone marrow of most, but not all cases associated with a shorter overall survival than AML with
shows abnormal eosinophils with coarse basophilic granules inv(16). Therefore, these cases should not be considered a
that may also be positive for naphthol-ASD-chloroacetate- variant of the AML-defining inv(16) [62].
esterase reaction. The abnormal granulation of eosinophils is
evident on bone marrow aspirate smears but usually cannot 13.3.4.5 Caution
be observed in section cut from paraffin-embedded biopsies. • The typical coarse basophilic granules of eosinophils may
By contrast, rare abnormal eosinophils may already be            be missed in sections of paraffin-embedded bone marrow
detected in some cases with a low percentage of blasts <20%      core biopsies.
that already carry a chromosome 16 abnormality and should • Eosinophils with abnormal basophilic granules are not
be diagnosed as overt AML (see case history).                    present in all cases of AML with inv(16)(p13.1q22) or
                                                                 t(16;16)(p13.1q22).
13.3.4.3 Immunophenotype                                     • Eosinophils may be increased in other AML subtypes,
Myeloid antigens (CD13, CD33, MPO), monocyte- for example, AML with t(8;21)(q22;q22.1) RUNX1-
associated markers (CD4, CD11b, CD14, CD64), and mark-           RUNX1T1.
13       Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies                                                 403
Case History 1: AML with inv(16)(p13.1q22) (CBFB/                         However, molecular studies revealed a CBFB/MYH11
MYH11) (Figs. 13.6 and 13.7)                                              translocation. Therefore, the diagnosis was revised to AML
A 29-year-old male was referred with pancytopenia and the                  with inv(16) despite the low percent of blasts. The patient
suspicion of a myelodysplastic syndrome. The CBC was:                      refused AML-type therapy. Five months later, peripheral leu-
WBC 2.5 × 109/L, Hb 9.1 g/dL, platelets 89 × 109/L. Differential           kocytosis with monocytosis and the presence of 24% blasts
count: 8% monocytes, 11% neutrophils including some                        and promonocytes developed. The bone marrow aspirate
pseudo-Pelger forms, 1% blast, 3% promonocytes, 4%                         smears and core biopsy contained blasts and precursors of
eosinophils, and 67% lymphocytes. The bone marrow aspi-                    myelopoiesis and monocytopoiesis. Numerous abnormal
rate smear revealed a hypocellular marrow with 16% blasts                  eosinophils, some of which contained abnormal purple-vio-
and scattered atypical hypergranulated eosinophils                         let granules, were observed (Fig. 13.7). The bone marrow
(Fig. 13.6). By immunophenotyping, 12% CD117-positive                      biopsy was moderately hypercellular and showed an increase
cells were present that co-expressed (>90%) CD13, CD38,                    in eosinophils, but the abnormal granulation was only evi-
CD45 lo, and MPO and were partially positive for CD34                      dent on the aspirate smears. The neoplastic population was
(35%), HLA-DR (35%), and CD33 (25%). The core biopsy                       positive for myeloperoxidase and partially expressed CD14,
was hypocellular and showed reduction of all three hemato-                 CD68, and CD34. Karyotyping, FISH, and RT-PCR analysis
poietic lineages with mild erythropoietic dysplasia.                       confirmed a CBFβ-rearrangement / inv(16)(p13.1q22) con-
Maturation of granulopoiesis was rarely observed. Blast                    sistent with the diagnosis of AML with inv(16)(p13.1q22).
cells accounted for about 15–16% of nucleated cells                        In addition, NGS using a myeloid panel revealed KRAS- and
(Fig. 13.6). The diagnosis was MDS-excess blasts 2.                        TET2 mutations.
a b
                                                               MG
                                                               M GG                                                                      MGG
 c                                                                         d
                                                               MGG
                                                               MGG
                                                               MG                                                                        MGG
                                                                                                                                         MGG
                                                                                                                                         MG
Fig. 13.6 AML with inv(16)(p13.1q22) and low blast count, case 1:         precursors with coarse granules are appreciated (a–d, MGG). (e–h) The
(a) Eosinophils with normal granulation and (b) some monocytes with       bone marrow biopsy is hypocellular and shows a relative predominance
abnormal nuclear lobation were detected in the peripheral blood smears.   of erythropoietic cells, diminished granulopoietic maturation, and rare
(c, d) Hypocellular bone marrow aspirate smears with (c) rare blasts      dysplastic megakaryocytes (e, f, Giemsa; g, h, NASDCL)
and groups of erythropoietic cells. (d) Scattered atypical eosinophil
404                                        13   Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies
e f
Giemsa Giemsa
g h
                                                       N
                                                       NA
                                                        AS
                                                         SDDCL
                                                            C
                                                       NASDCL                                                                  NASDCL
                                                                                                                               N
                                                                                                                               NA
                                                                                                                                AS
                                                                                                                                ASD
                                                                                                                                 SDDCL
                                                                                                                                    CL
a b
MGG MGG
Fig. 13.7 AML with inv(16)(p13.1q22), case 1: (a, b) Sequential bone   stained sections. (d) Eosinophils show an aberrant Naphthol-ASD-
marrow smear contains among immature myelomonocytic precursors         chloroacetate-esterase positivity. The neoplastic myelomonocytic cells
and blasts numerous abnormal eosinophils with pleomorphic coarse       express myeloperoxidase (e). Subpopulations express CD34 (f), CD14
often purple-violet granules (MGG). (c–h) Bone marrow core biopsy.    (g), and CD68 (h)
(c) The abnormal granulation of eosinophils is not evident on Giemsa
13   Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies       405
c d
Giemsa NASDCL
e f
MPO CD34
g h
                                                        CD14
                                                        CD1
                                                        CD 14
                                                            4                                CD68
                                                                                             C
                                                                                             CD
                                                                                              D68
                                                                                                68
Case History 2: AML with CBFB/MYH11 inv(16)                               to a diffuse infiltration by blasts and immature myelomono-
(p13.1q22) CBFB/MYH11 (Fig. 13.8)                                         cytic cells (Fig. 13.8). Eosinophils were increased; however,
A 43-year-old woman presented to the hospital with shortness              no abnormal purple granules were observed on the sections. A
of breath, weakness, and enlarged cervical lymph nodes. CBC               bone marrow aspirate was not available due to a dry tap result-
at presentation: WBC 58 × 109/L, Hb 8.6 g/dL, platelets                   ing from packed marrow. The karyotype was 46,XX,inv(16)
39 × 109/L. Differential count: 46% monocytes, 10% neutro-                (p13.1 q22) [11]. FISH showed a CBFβ-rearrangement. A
phils, 24% promonocytes and monoblasts, 1% eosinophils,                   CBFB-MYH11 fusion transcript was detected by
and 19% lymphocytes (Fig. 13.8). The blasts were positive                 RT-PCR. Further molecular analysis using a next-generation
for sCD11c, sCD14, sCD64, sCD11b, sCD33, sCD13,                           myeloid panel revealed an additional NRAS mutation.
sCD15, and HLA-DR. The core biopsy was hypercellular due                     The diagnosis was AML with inv(16)(p13.1q22).
a b
                                                               MG
                                                               MG
                                                                GG
                                                                 G                                                                        MPO
c d
                                                            Giemsa                                                                    Giem
                                                                                                                                      Giemsa
                                                                                                                                           sa
                                                                                                                                      Giemsa
Fig. 13.8 AML with inv(16)(p13.1q22), case 2: (a, b) Blood smears         blastic and monoblastic features. Eosinophil precursors are present (d);
with (a, MGG) atypical monocytes, granulated myelomonocytic cells,        however, abnormal granulation is not prominent on the sections (c, d,
and (b) myeloperoxidase-positive blasts. (c–h) The bone marrow core       Giemsa). The neoplastic population expresses (e) myeloperoxidase, (f)
biopsy is hypercellular and diffusely infiltrated by blasts with myelo-   CD33, (g) partially CD68, and (h) strongly lysozyme
13   Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies         407
e f
MPO CD33
g h
                                                        CD68
                                                        CD
                                                        CD68
                                                           68                              Lysozyme
13.3.5 A
        cute Myeloid Leukemia with Translocation              cation [63, 66]. Basophilia and multilineage dysplasia
       t(6;9)(p23;q34.1); DEK-NUP214                           especially of the megakaryocytic lineage may be seen.
Case History: AML t(6;9);DEK-NUP214 (Fig. 13.9)                      marrow biopsy was infiltrated by myeloid blasts (about 60%)
A 72-year-old male patient was admitted with leukocytosis            and showed increased highly dysplastic megakaryocytes,
(WBC 20 × 109/L), anemia (Hb 7.9 g/dL), and thrombocyto-             dyserythropoiesis, and some residual granulocytic precur-
sis (534 × 109/L). Peripheral blood contained about 80%              sors. Karyotyping revealed t(6;9)(p23;q34.1).
blasts that were positive for CD13, CD33, CD38, HLA-DR,                 The diagnosis was AML with translocation
CD117, and partially myeloperoxidase (55%). The bone                 t(6;9);DEK-NUP214.
a b
                                                               HE
                                                               HE                                                                    HE
c d
HE NASDCL
Fig. 13.9 Acute myeloid leukemia with translocation t(6;9);DEK-     present. Blast represented about 60% of nucleated cells and expressed
NUP214. (a–c, H&E) The bone marrow core biopsy was hypercellular     (e) myeloperoxidase and (f) partially CD68. (g) CD71 immunolabeling
and showed prominent megakaryocytic dysplasia with numerous          highlighted the presence of dysplastic erythropoietic precursors. (h)
micromegakaryocytes and megakaryocytes with mono- or bilobated       CD34+ blast were rare
nuclei. (d) Some NASDCL+ granulocytes and myeloid precursors were
410                     13   Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies
e f
MPO CD68
g h
CD71 CD34
13.3.6 A
        ML with Translocation t(9;11)                              lymphoblastic leukemia (ALL). It has been suggested that
       (p21.3;q23.3); KMT2A-MLLT3                                    disordered methylation is a critical leukemogenic event in
                                                                     these neoplasms. Important for the leukemogenic function
AML with translocation t(9;11)(p21.3;q23.3) resulting in the         of KMT2A is the DOT1L histone methyltransferase that
KMT2A-MLLT3 fusion protein represents a distinct entity in the       may be a target by specific inhibitors [70]. An age-depen-
2016 WHO classification [11]. The translocation t(9;11) occurs       dent breakpoint shift with breakpoints localizing within
in both de novo and therapy-related AML. Epipodophyllotoxins         MLL intron 11 has been reported to be associated with acute
like etoposide have been associated with a short interval between    lymphoblastic leukemia and younger patients, while break-
treatment and development of t-AML, with fusion oncogenes            points in MLL intron 9 predominate in AML or older patients
like KMT2A/MLL-MLLT3 [68]. However, only the de novo                 [71]. In about 40% of AML with t(9;11) a MECOM (EVI1)
cases are included in this entity while cases of secondary AML       overexpression is found. MECOM (EVI1) has been shown
with this translocation are classified as therapy-related myeloid    to define a poor prognostic subset of MLL-rearranged AML
neoplasm [66, 68].                                                   [72]. MECOM-negative cases may harbor a gain of chromo-
                                                                     some 8 [11].
13.3.6.1     Epidemiology and Clinical                                  Numerous variant KMT2A translocations have been
              Presentation                                           reported involving genes such as MLLT1, MLLT3; MLLT4 or
Rearrangements of the human MLL/KMT2A gene occur in                  MLLT4, and other genes, resulting predominantly in ALL or
infant, pediatric, and adult AML patients. The translocation         AML [11, 73]. MLL-rearranged pediatric AML is a heteroge-
t(9;11) resulting from KMT2A gene translocation with the             neous disease, and prognosis depends on various factors, for
(p22;q23) breakpoint is more frequent in the pediatric age           example, translocation partner, age, WBC, and additional
group where it occurs in up to 12% of AML cases while it is          cytogenetic aberrations [69].
reported in only about 2% of adult AML patients [11, 69].                There is evidence of a complex “MLL recombinome”
   Patients may present with disseminated intravascular              associated with different hematologic malignancies, and in
coagulation and/or extramedullary tissue infiltrates/myeloid         particular with acute leukemia (ALL and AML) [71].
sarcoma [11].                                                        According to the 2016 WHO classification, hematopoietic
                                                                     neoplasms associated with 11q23 translocation but without
13.3.6.2 Morphology                                                 KMT2A-rearrangement (11q23+/KMT2A−) are not included
Leukemic populations harboring t(9;11)(p21.3;q23.3);                 in this AML entity (Case history 2. Fig. 13.11).
KMT2A-MLLT3 are generally composed of neoplastic cells
with features of promonocytes and monoblasts.                       13.3.6.5 Caution
                                                                    • AML cases presenting with morphologic and phenotypic
13.3.6.3 Immunophenotype                                             features of monocytic/monoblastic leukemia should not be
Adult AML cases frequently express monocytic markers such             classified as AML, NOS without an analysis of the genetic
as CD14, CD4, CD11b, CD11c, CD64, CD36, and lysozyme.                 profile, especially a t(9;11)(p21.3;q23.3); KMT2A-MLLT3.
Expression of CD34, CD117, and CD56 is variable. Pediatric          • Cases with variant translocations of the mixed-lineage leuke-
cases are reported to strongly express CD33, CD56, CD4, and           mia (MLL) gene to other fusion partners do not fit into this
HLA-DR while CD34, CD13, and CD14 are usually low [11].               category of AML with t(11;16)(q23;q12); KMT2A-MLLT3.
                                                                    • Cases of AML with t(11;16)(q23;q12)); KMT2A-MLLT3
13.3.6.4 Genetics                                                    developing secondary to cytotoxic chemotherapy and/or
Chromosome 11q23 translocations involving the lysine                  radiation therapy for malignant tumors are not included in
(K)-specific methyltransferase 2A gene (KMT2A, previ-                 the entity but have to be classified as therapy-related
ously MLL) have been well characterized in AML and acute              neoplasms.
412                                          13   Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies
Case History 1: AML with t(9;11)(p21.3;q23.3);                           CD2, CD3 i.c., CD5, CD7, CD19, CD56, CD65, CD79a,
KMT2A-MLLT3 (Fig. 13.10)                                                and CD117. FISH analysis revealed a MLL (11q23)-rear-
The patient was a 54-year-old woman presenting with swell-               rangement. The karyotype was 46, XX, t(9;11)(p22;q23),
ing of the gingiva, leukocytosis (WBC 34 × 109/L), anemia                MLLT3-KMT2A. No additional mutations were detected.
(Hb 7.3 g/dL), and thrombocytopenia (platelets 61 × 109/L).                 The bone marrow core biopsy was infiltrated by blasts
The peripheral blood smear showed 2% band forms, 7%                      representing more than 90% of nucleated cells. The pheno-
neutrophils, 12% lymphocytes, and 79% promonocytes and                   type was consistent with a monoblastic differentiation high-
monoblasts that were positive for CD4, CD11b, CD11c,                     lighted by a strong expression of CD68 (Fig. 13.10). The
CD13, CD14, CD15, CD33, CD38, CD45, CD64, CD68,                          gingiva was infiltrated by the same promonocytic and mono-
lysozyme, and HLA-DR. A small percentage expressed                       blastic population. The diagnosis was de novo AML with
CD34 and/or myeloperoxidase. The blasts were negative for                t(9;11)(p21.3;q23.3); KMT2A-MLLT3.
      a
                                                                         b
                                                           Giemsa                                                                    Giemsa
c
                                                                         d
                                                               MPO                                                                       MPO
                                                                                                                                         MP
                                                                                                                                         M PO
Fig. 13.10 AML with t(9;11)(p21.3;q23.3); KMT2A-MLLT3. The bone          and gray cytoplasm (a, b, Giemsa). A minority of blasts expressed myelo-
marrow core biopsy was diffusely infiltrated by a promonocytes and       peroxidase (c, d) while the large majority was positive for CD68 (e, f).
monoblasts with round or slightly indented nuclei, prominent nucleoli,   Endothelial cells and rare blasts were positive for CD34 (g, h)
13   Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies     413
e f
                                                        CD68                               CD68
g                                                                 h
                                                        CD
                                                        CD3
                                                          34
                                                        CD344                              CD34
                                                                                           CD
                                                                                           CD3
                                                                                             34
                                                                                              4
Case History 2: AML with Variant KMT2A                                     moderately hypercellular with a predominance of CD117,
Translocation (Fig. 13.11)                                                 CD33, and MPO expressing blasts. The residual erythropoi-
A 47-year-old female patient was admitted with pancytope-                  etic islands and megakaryocytes clearly exhibited dysplastic
nia and a suspected myelodysplastic syndrome. CBC at pre-                  features (Fig. 13.11).
sentation: WBC 0.4 × 109/L, Hb 3.8 g/dL, platelets                            FISH analysis revealed a MLL (11q23)-rearrangement.
8 × 109/L. The peripheral blood aspirate smears were hypo-                 The karyotype was 46, XX t(11;16)(q23;q12) consistent
cellular and contained 2% blasts, 16% band forms, 2% baso-                 with a reciprocal translocation between a long arm of chro-
phils, 3% monocytes, and 77% lymphocytes.                                  mosome 11 with a long arm of chromosome 16. The diagno-
    The bone marrow was infiltrated by CD117-positive                      sis was AML with a KMT2A (MLL)-rearrangement with an
blasts co-expressing CD13, CD33, MPO, HLA-DR, and par-                     unknown fusion partner. This case was therefore not included
tially CD34. Blasts were negative for CD7, CD15, CD56,                     in the 2016 WHO entity “AML with t(9;11)(p21.3;q23.3);
CD79a, and lysozyme. The bone marrow core biopsy was                       KMT2A-MLLT3”.
a b
Giemsa Giemsa
c d
                                                           NASDCL                                                                     MP
                                                                                                                                      MPO
                                                                                                                                       PO
Fig. 13.11 AML with t(11;16)(q23;q12). The bone marrow core                CD117 (f). Residual erythropoiesis shows a disturbed maturation (g,
biopsy is infiltrated by myeloid blasts with a slightly granulated cyto-   CD71). Megakaryocytes are clearly dysplastic and predominantly cor-
plasm (a, b, Giemsa). Neoplastic cells are partially positive for          respond to micromegakaryocytes often arranged in small clusters (h,
NASDCL (c) and strongly express myeloperoxidase (d), CD33 (e), and         CD61)
13   Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies      415
e f
CD33 CD117
g h
                                                        CD71
                                                        CD
                                                        CD7
                                                          711                              CD61
                                                                                           CD
                                                                                           CD6
                                                                                             61
                                                                                              1
13.3.7 A
        ML (Megakaryoblastic) with                             be positive for CD33 and CD13 while CD34, CD45, and
       Translocation t(1;22)(p13.3;q13.1);                      HLA-DR are often negative [11, 74]. An immunohisto-
       RBM15-MKL1                                               chemical study including different subsets of AMKL has
                                                                shown that CD42b can reliably be used as a solitary first-
This extremely rare AML entity is generally associated with     line marker for blasts of megakaryocytic lineage, whereas
morphologic and immunophenotypic features similar to            CD61 may be reserved for infrequent cases that are CD42b-
acute megakaryoblastic leukemia (AMKL), a subtype of            negative [76].
AML, NOS.
                                                                13.3.7.4 Genetics
13.3.7.1 Epidemiology and Clinical Features                    Non-Down syndrome AMKL is characterized by chimeric
AML with translocation t(1;22)(p13.3;q13.1); RBM15-            oncogenes including RBM15-MKL1, CBFA2T3-GLIS2,
MKL1 represents <1% of all AML cases and develops in            NUP98-KDM5A, and MLL gene rearrangements. CBFA2T3-
infants and young children ≤3 years and may even be con-        GLIS2 is the most frequent chimeric oncogene identified to
genital. Symptoms at diagnosis include organomegaly,            date in this subset of patients and confers a poor prognosis
especially hepatosplenomegaly, thrombocytopenia, and            [74]. However, in the 2016 WHO classification, only cases
mild elevated WBC [11]. The outcome of non-Down syn-            harboring the reciprocal chromosomal translocation t(1;22)
drome AMKL is generally poor, with lower event-free             (p13;q13) that originates the fusion oncogene RBM15-
survival than DS-AMKL and pediatric AML [74].                   MKL1 are included as a distinct entity [11]. The t(1;22)
According to a multi-center prospective study, pediatric        (p13;q13) translocation was first described in cohort of 6/252
patients with AMKL (excluding Down syndrome) had a              children with AML all of whom were <1 year of age [77].
significantly poorer outcome than other childhood AML           MKL1 is a transcriptional coactivator for serum response
subgroups [75].                                                 factor (SRF), a transcription factor that regulates the expres-
                                                                sion of genes involved in cell growth, proliferation, and dif-
13.3.7.2 Morphology                                            ferentiation, as well as genes that control the actin
The blasts may be rare in peripheral blood and also bone        cytoskeleton. The fusion of MKL1 to RBM15 deregulates the
marrow smears due to bone marrow fibrosis resulting in a        normal intracellular localization of MKL1 such that it
dry tap. Blasts show large nuclei with dense chromatin and      becomes constitutively localized to the nucleus, resulting in
an undifferentiated cytoplasm with irregular cytoplasmic        SRF activation even in the absence of stimuli [74].
margins and so-called blebs. The bone marrow generally
reveals a reticulin to collagen fibrosis leading to difficult   13.3.7.5 Caution
marrow aspirations and a falsely decreased blast count.         • In infants, a clonal proliferation of megakaryoblasts may
Therefore, a bone marrow core biopsy may be mandatory for         be observed in Down syndrome-associated transient
further diagnostic procedures including the evaluation of the     abnormal myeloproliferative disorder (TMD) that may
blast percentages. For the diagnosis of AML blasts should be      also show myeloblastic and erythroblastic components.
≥20% with the exception of cases with extramedullary tissue          TMD may progress to myeloid leukemia (ML-DS)
infiltrates/myeloid sarcoma.                                      with morphologic features reminiscent of AMKL.
                                                                     However, in contrast to AMKL, TMD and ML-DS
13.3.7.3 Immunophenotype                                         harbor mutations in the hematopoietic transcription fac-
Blast cells express platelet-specific surface glycoprotein        tor GATA1 [78].
such as CD41, CD42b and CD61 and are negative for               • In adult patients, most cases diagnosed with morphologic
myeloperoxidase, TdT, and lymphoid markers. They may              features of AMKL belong to the AML, NOS category.
13    Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies                                                 417
Case History: AML with Translocation t(1;22)                            partially CD33 but were negative for myeloperoxidase,
(p13.3;q13.1); RBM15-MKL1 (Fig. 13.12)                                  CD34, CD117, and glycophorin C consistent with a mega-
A 6-month-old male baby was diagnosed with an anterior                  karyoblastic phenotype. The reticulin fiber network was
mediastinal mass. CBC at presentation: WBC 16 × 109/L, Hb               increased (fibrosis grade 1). A mediastinal biopsy revealed
9.1 g/dL, platelets 80 × 109/L. The peripheral blood smears             an extramedullary tumor forming tissue infiltration by the
revealed 52% undifferentiated myeloperoxidase-negative                  same blast cell population (Fig. 13.12).
blasts with prominent cytoplasmic blebs that represented                   Cytogenetic and molecular analyses provided evidence of an
85% of bone marrow cells in the aspirate smears (Fig. 13.12).           AML with translocation t(1;22)(p13.3;q13.1); RBM15-MKL1
Blasts expressed CD42b, CD61, von Willebrand factor, and                associated with a myeloid sarcoma at initial presentation.
a b
                                                             MGG                                                                      MGG
 c                                                                      d
                                                               PAS                                                                   CD34
                                                                                                                                     C
                                                                                                                                     CD
                                                                                                                                      D334
                                                                                                                                         4
Fig. 13.12 AML (megakaryoblastic) with translocation t(1;22)            that are negative for CD34 (d) but partially positive for CD33 (e) and
(p13.3;q13.1); RBM15-MKL1. Large undifferentiated blasts with           strongly express CD61 (f). The mediastinal tumor is composed of a
prominent cytoplasmic blebs are observed (a) in the peripheral blood    phenotypically identical neoplastic population consistent with a
and (b) the bone marrow aspirate smears (MGG). The bone marrow          myeloid sarcoma (g, h; PAS)
core biopsy (c–f) is infiltrated by large polymorphic blasts (c, PAS)
418                      13   Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies
e f
CD33 CD61
g h
                                          PAS
                                          PAS
                                          PA                                                               PAS
                                                                                                           PA
                                                                                                           PAS
                                                                                                            AS
13.3.8 AML with Translocation                                    reminiscent of CML such as small megakaryocytes and/or
        t(9;22);BCR-ABL1                                          pseudo-Gaucher cells.
Case History (Fig. 13.13)                                                92% of peripheral blood cells. An identical population was
A 59-year-old male patient was initially admitted to the hospi-          present in bone marrow aspirate smears and in the trephine
tal with a suspected diagnosed of AML, NOS without matura-               bone marrow biopsy (Fig. 13.13). Erythro- and granulocyto-
tion. No previous history of a hematologic or non-hematologic            poiesis were significantly reduced. The marrow spaces were
neoplasm was reported. Clinical examination revealed no                  hypercellular. However, some fat cells were still present.
splenomegaly. CBC at presentation: WBC 33 × 109/L, Hb                    Karyotyping revealed a translocation t(9;22)(q34.1;q11.2).
6.8 g/dL, platelets 74 × 109/L. Myeloid blasts expressing                Molecular analysis provided evidence of a p210 fusion tran-
CD13, CD33, CD34, CD7, and partially MPO represented                     script. The diagnosis was AML t(9;22);BCR-ABL1-positive.
a b
                                                          Giemsa                                                                      Giemsa
 c                                                                       d
Giemsa NASDCL
Fig. 13.13 AML with translocation t(9;22)/BCR-ABL1-positive. (a–         addition to residual granulopoietic cells, a minority of blasts expressed
h) The bone marrow biopsy was hypercellular, but in contrast to most     MPO. (g, h). CD34 was strongly expressed in the total leukemic popu-
cases with CML BP still contained some fat cells. Blasts were medium-   lation. No residual dwarf megakaryocytes or macrophages with fea-
sized without significant maturation (a–c; Giemsa). (d) Scattered        tures of pseudo-Gaucher cells that may be present in CML BP were
NASDCL+ myeloid cells represented residual granulopoiesis. (e, f) In     detected
13   Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies     421
e f
MPO MPO
g h
CD34 CD34
13.4     AML with Gene Mutations                                  used to assess residual blasts after chemotherapy. Blasts gen-
                                                                   erally express strongly CD33 and often monocytic markers
This category of the revised 2016 WHO classification               such as CD14, CD36, and CD64 but are mostly negative for
includes the frequently diagnosed subtype AML with                 CD34. However, CD34 positivity may occur. A subpopula-
mutated NPM1, AML with biallelic mutation of CEBPA, and            tion expressing stem cell markers may be detected by flow.
the provisional entity AML with mutated RUNX1.                     The expression of CD34, CD25, CD123, and CD99 has been
                                                                   associated with FLT3-ITD co-mutations [11].
Case History 1: AML with Mutated NPM1 and FLT3                            tion and a FLT3-ITD co-mutation (ratio 0.6). The peripheral
ITD Co-mutation (Fig. 13.14)                                              blood smear contained dysplastic myeloid cells with pseudo-
The patient was a 42-year-old woman who reported inguinal                 Pelger-like features and multiple blasts with nuclear cup- or
lymph node swelling and abnormal uterine bleeding symp-                   fish-mouth-like nuclear invaginations. The trephine bone
toms. At presentation, the CBC was: WBC 8 × 109/L, Hb                     marrow biopsy was infiltrated by blasts with monocytoid fea-
6.1 g/dL, platelets 35 × 109/L. Myeloid blasts accounted for              tures strongly expressing CD33 and clearly showing a cyto-
80% of peripheral blood and 65% of nucleated bone marrow                  plasmic translocation of the NPM1 protein. In addition, there
cells. Blasts were positive (>90%) for CD13, CD33, CD38,                  was evidence of prominent erythroid and megakaryocytic
CD45 lo, CD64, and HLA-DR; and partially positive for                     dysplasia. Despite multilineage dysplasia, the correct diag-
CD11c (90%), MPO (50%), and CD34 (20%). The karyo-                        nosis was not AML with MDS-related changes (MRC) but
type was normal. Molecular analysis revealed a NPM1 muta-                 AML with mutated NPM1 and co-mutation of FLT3-ITD.
a b
                                                               MG
                                                                GG                                                                 Giemsa
 c                                                                        d
                                                            Gie
                                                            Giemmsa
                                                                  a                                                              NASDCL
                                                                                                                                 NA
                                                                                                                                 NAS
                                                                                                                                   SD
                                                                                                                                    DCCL
                                                                                                                                       L
Fig. 13.14 AML with mutated NPM1 and FLT3-ITD co-mutation                 karyocytic dysplasia with clusters of atypical small megakaryocytes
(case 1). Cup-like nuclear inclusions in blast cells were a hallmark of   negative for NASDCL (d) but highlighted by CD61 immunohistochem-
peripheral blood and bone marrow aspirate smears (a, MGG).                istry (f). Large erythropoietic island with disturbed maturation (g,
Infiltration of the bone marrow core biopsy by blasts with prominent      CD71). Aberrant cytoplasmic NPM1 staining in addition to nuclear
nucleoli and a basophilic cytoplasm (b, c, Giemsa) that were negative     immunolabeling (h) as a typical feature of this AML entity
for NASDCL (d) but strongly expressed CD33 (e). Note marked mega-
424                      13   Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies
e f
CD33 CD61
g h
                                         CD71
                                         CD
                                         CD71
                                            71                                                           NPM1
Case History: Case 2. AML with Mutated NPM1 and                            Cytochemical stains performed on bone marrow smears also
FLT3 Co-mutation (Fig. 13.15)                                              showed a MPO-positive blast subpopulation (Fig. 13.15).
A 58-year-old woman was admitted because of weakness,                      The bone marrow biopsy was diffusely infiltrated by blasts
dyspnea, fever, and weight loss developing during the last 3               with myelomonocytic features. The karyotype was normal.
weeks. At clinical examination, her skin and mucosa were                   The initial diagnosis was acute myelomonocytic leukemia.
extremely pale. The CBC was: WBC 49 × 109/L, Hb 4.2 g/                     However, immunohistochemistry performed on bone mar-
dL, platelets 116 × 109/L. The LDH was 634 U/L. Peripheral                 row sections revealed aberrant cytoplasmic NPM1 expres-
blood smears showed 78% blasts, 2% metamyelocytes, 2%                      sion suggestive of a NPM1 mutation. The NPM1 mutation
neutrophils, 2% eosinophils, 2% basophils, and 14% lym-                    (exon 12) was confirmed by molecular analysis that reported
phocytes. Blasts were positive for CD33, CD117, CD45 lo,                   an additional FLT3 ITD mutation (ratio 0.65). Retrospective
and CD64 and partially positive for CD11c (90%), CD13                      evaluation of cytologic specimens revealed some blasts with
(90%), CD38 (90%), CD7 (25%), CD86 (25%), CD11b                            cup-like nuclear inclusions (Fig. 13.4). The final diagnosis
(20%), CD14 (15%), Lysozyme (15%), and MPO (11%).                          was AML with mutated NPM1 and FLT3-ITD.
a b
MGG MGG
c d
MGG Giemsa
Fig. 13.15 AML with mutated NPM1 and FLT3 co-mutation, case 2.             hypercellular bone marrow core biopsy by blasts with basophilic cyto-
Peripheral blood (a) and bone marrow smears (b, c) with abundant blasts    plasm (d, e, Giemsa) intermingled with macrophages containing cyto-
with a basophilic cytoplasm and round or slightly indented nuclei with     plasmic debris and some residual megakaryocytes (e). Blasts strongly
some cup-like inclusions (a, b, MGG). MPO positivity was observed in       expressed CD33 (f), rarely CD14 (g), and clearly showed an aberrant
a subpopulation (c, MPO cytochemistry). Dense infiltration of the highly   nuclear and cytoplasmic NPM1 immunolabeling (h)
426                      13   Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies
e f
Giemsa CD33
g h
                                         CD14
                                         CD14
                                         CD14                                                            NPM1
Case History: Case 3. AML with Mutated NPM1,                              tained necrotic cells with remnants of blasts and eosinophils
DNMT3A, IDH2, and Charcot Crystals (Fig. 13.16)                           including typical Charcot crystals. Some spaces were occu-
A 57-year-old male patient presented with severe multifocal               pied by a loose fibrotic stroma. Molecular analysis revealed
bone pain. The CBC was: WBC 64 × 109/L, Hb 7.9 g/dL,                      mutation of NPM1, DNMT3A, and IDH2. The trabecular
platelets 48 × 109/L. The peripheral blood contained 4%                   bone trabecel showed focal zones of absorption.
eosinophils and ≥90% blasts that were positive for CD 117,                   The diagnosis was NPM1-mutated AML with DNMT3A,
CD13, CD33, and CD38; and partially positive for CD11c                    IDH2 co-mutation, and Charcot crystals.
(85%), HLA-DR (75%), CD45 lo (50%), CD7 (35%), and                           Caution: Charcot crystals may not only occur in second-
MPO (30%). No bone marrow aspirate could be obtained                      ary eosinophilia in allergic disorders or hypereosinophilic
(dry tap). The trephine bone marrow biopsy showed hyper-                  syndromes but also in AML with an eosinophilic component
celluar areas infiltrated by myeloblasts intermingled with                [96]. Especially, an association with NPM1-mutated AML
numerous eosinophils. Adjacent bone marrow spaces con-                    has been described [97].
a b
Giemsa Giemsa
c d
Giemsa Giemsa
Fig. 13.16 AML with mutated NPM1, DNMT3A, IDH2 and Charcot                (g, Giemsa; h, NASDCL) marrow spaces. Note presence of abundant
cristals Pre-treatment bone marrow core biopsy showing partially          eosinophils (c) and typical Charcot cristals especially in areas of spon-
hypercellular (a–e, Giemsa; f, NASDCL), partially hypocellular fibrotic   taneous necrosis (d–e)
428                      13   Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies
e f
Giemsa NASDCL
g h
                                      Giem
                                      Giem
                                      Gi ms
                                          sa
                                      Giemsaa                                                        NASDCL
                                                                                                     N
                                                                                                     NA
                                                                                                      ASD
                                                                                                       SDCCL
                                                                                                           L
Case History 1 (Fig. 13.17)                                          The bone marrow biopsy was hypercellular and infiltrated by
The initial symptom of this 37-year-old male patient was the         CD33-positive blasts partially positive for NASDCL and
development of cutaneous and mucosal hemorrhage. The                 rarely for CD14. No background myelodysplastic changes
CBC was: WBC 18 × 109/L, Hb 10.6 g/dL, platelets                     were observed. The karyotype was normal. Mutational anal-
21 × 109/L. Peripheral blood and bone marrow showed 85%              ysis revealed a biallelic CEBPA mutation. Therefore, the
blasts mostly positive for myeloperoxidase and negative for          case was assigned to the category AML with biallelic muta-
nonspecific esterase suggestive of an AML with maturation.           tion of CEBPA.
a b
Giemsa Giemsa
c d
Giemsa NASDCL
Fig. 13.17 AML with biallelic mutation of CEBPA. The trephine bone   cells and showed features of AML with maturation and were partially
marrow biopsy was hypercellular with some residual fat cells (a–c,   positive for NASDCL (d). In addition, CD33 was expressed in the
Giemsa). Blasts accounted for about 90% of nucleated bone marrow     majority of blasts (e, f) and CD34 in a subpopulation (g, h)
13   Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies     431
e f
CD33 CD33
g h
                                                        CD34
                                                        CD3
                                                        CD344                              CD34
                                                                                           CD
                                                                                           CD3
                                                                                             34
                                                                                              4
Case History 2 (Fig. 13.18)                                                 The karyotype was normal. Molecular analysis revealed a
A 47-year-old woman was admitted with widespread cutane-                 biallelic CEBPA mutation in addition to a GATA2 zinc finger
ous hemorrhage. The CBC was: WBC 80 ×109/L, Hb 7.2 g/                    (ZF)1 mutation. The diagnosis was normal karyotype AML
dL, platelets 13 × 109/L. The bone marrow aspirate contained             with biallelic CEBPA-mutation with concomitant GATA2
85% blasts expressing myeloperoxidase, CD7, CD38,                        mutation.
CD117, and HLA-DR and partially positive for CD33, CD13
(50%), CD15 (45%), and CD86 (40%).
a b
MGG MGG
c d
Giemsa Giemsa
e f
NASDCL MPO
Fig. 13.18 AML with biallelic mutation of CEBPA and of GATA2. (a,        marrow spaces diffusely infiltrated ba blasts that were largely negative
b) Peripheral blood (a) and bone marrow aspirate smears (b) containing   for naphthol-ASD-chloroacetate-esterase (e) but strongly expressed
medium-sized blasts accounting for about 80% of nucleated bone mar-      myeloperoxidase (f), CD7 (g), and CD117 (h)
row cells. (c, d) Trephine bone marrow biopsy showing hypercellular
13   Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies      433
g h
CD7 CD117
Case History                                                               positive for CD34, CD13, CD38, HLA DR, and CD117 and
A 68-year-old man consulted his physician because of a                     partially positive for CD7 (68%)and CD56. Morphology
perianal abscess pancytopenia. The CBC was: 1.2 × 109/L,                   and immunophenotype were suggestive of an AML with
Hb 6.8 g/dL, platelets 102 × 109/L. The differential count                 minimal differentiation. The karyotype was normal.
revealed 26% MPO-negative nongranulated medium-sized                       Molecular analysis revealed a monoallelic RUNX1 muta-
MPO-negative blasts, 2% metamyelocytes, 14% neutro-                        tion and a KMT2A partial tandem duplication (KMT2A-
phils, 1% eosinophils, 46% lymphocytes, and 11% smudged                    PTD). The diagnosis was AML with mutated RUNX1
cells. The bone marrow contained 70% blasts that were                      (Fig. 13.19).
a b
                                                                   HE                                                                  HE
 c                                                                         d
CD34 CD56
Fig. 13.19 AML with mutated RUNX1. The bone marrow biopsy was              for MPO but strongly expressed CD34 (c). A majority of blasts was
hypercellular but still contained fat cells (a, b, HE). Immature appear-   positive for CD56 (d) and CD7 (e, f). NASDCL reaction (g) and MPO
ing blasts represented about 70% of nucleated cells and were negative      (h) immunohistochemistry marked residual granulopoietic cells
436                      13   Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies
e f
                                          CD7                                                              CD7
 g                                                 h
NASDCL MPO
13.5     AML with Myelodysplasia-Related                        [108]. In this analysis, only megakaryocyte morphology
          Changes (AML-MRC)                                      showed significant associations with mutation pattern or
                                                                 outcome. The presence of megakaryocytes with separated
Acute myeloid leukemia with myelodysplasia-related               lobes was correlated with cohesin pathway and NPM1
changes (AML-MRC) is defined by the presence of                  mutations but had no influence on the prognosis. The pres-
≥20% peripheral blood or bone marrow blasts associated           ence of micromegakaryocytes was associated with poor
with morphologic features of multilineage dysplasia              outcome, but did not correlate with any mutations [108].
(MLD), and/or myelodysplastic syndrome (MDS)-related             According to a previous study that did, however, not con-
cytogenetics, and/or previous MDS or MDS/MPN [12].               sider the mutational status, multilineage dysplasia had no
Patients with AML-MRC are likely to have high-risk              impact on outcome, but the presence of micromegakaryo-
cytogenetics, leading to poor therapy response and prog-         cytes and hypogranulated myeloid cells was associated
nosis [12, 106, 107].                                            with adverse event free survival [109]. However, identifica-
                                                                 tion of morphologic dysplasia among observers is not
                                                                 always consistent, and there is morphologic overlap with
13.5.1 Epidemiology                                             other leukemic disorders such as acute erythroleukemia
                                                                 [110]. In AML-MRC, erythropoiesis may be strikingly
AML-MRC is predominantly diagnosed in elderly patients           expanded and represent more than half of nucleated bone
and represents up to 24–35% of all AML cases. This subtype       marrow cells [111]. These cases with ≥50% but <80% ery-
is rare in the pediatric age group [12].                         throid precursors and ≥20% or more blasts should be clas-
                                                                 sified as AML-MRC and not as erythroleukemia, erythroid/
                                                                 myeloid type [12, 112].
13.5.2 Morphology
Case History 1 (Figs. 13.20 and 13.21)                                 eythropoiesis represented about 45% of nucleated cells,
A 74-year-old woman had been healthy and active until 3                showed megaloblastoid changes, maturation defects, cyto-
weeks before consulting her physician because of weak-                 plasmic vacuolization and abnormal nuclear shape.
ness. The CBC was: BC 2.3 × 109/L, Hb 7.2 g/dL, platelets              Megakaryopoiesis was decreased and showed small atypi-
25 × 109/L. The peripheral blood showed dysplastic neutro-             cal cells. Residual granulopoietic cells were rare and dys-
phils with abnormal nuclear lobation and 17% MPO-                      plastic. By FISH analysis, a del(5q31/5q33), a monosomy
positive blasts (Fig. 13.19). Bone marrow smears contained             16, and a monosomy 17 were detected. Molecular analysis
23% myeloid blasts expressing CD34, CD33, and partially                revealed TP53, DNMT3A, and IKZF1 mutations. The diag-
MPO. Bone marrow smears showed a prominent back-                       nosis was AML with myelodysplasia-        related changes
ground dysplasia of more than 50% of cells. Dysplastic                 (AML-MRC).
a b
                                                             M G
                                                             MG                                                                     MGG
c d
                                                             MGG
                                                             MG                                                                     MGG
                                                                                                                                    M
                                                                                                                                    MGGG
Fig. 13.20 AML with myelodysplasia-related changes (AML-MRC).          with a markedly increased megaloblastoid erythroid lineage with cyto-
(a, b, MGG) Peripheral blood smears showing hypogranular dysplastic    plasmic vacuoles, binucleated erythroblasts, inappropriate nuclear
neutrophils (a) and rare blasts (b). (c–f, MGG) Bone marrow aspirate   lobation of the granulocytic lineage, and scattered myeloblasts
440                                          13   Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies
e f
MGG MGG
a b
HE PB
c d
HE MPO
Fig. 13.21 AML with myelodysplasia-related changes (AML-                  scattered MPO-positive immature myeloid cells (d), small dysplastic
MRC). The bone marrow core biopsy is hypercellular (a, HE), shows         megakaryocytes (h, CD61; also present in b, d), and CD34-positive
increased hemosiderin content of macrophages (b, Prussian blue            blasts (>20%, g)
stain), expansion of megaloblastoid erythropoiesis (c, HE; e, f, CD71),
13   Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies     441
e f
CD71 CD71
g h
                                                        CD34
                                                        CD
                                                        CD34
                                                           34                              CD61
                                                                                           C
                                                                                           CD
                                                                                            D661
                                                                                               1
13.6     Therapy-Related Myeloid Neoplasms                      amounts of blasts. Fibrosis may be present. t-AML following
                                                                 treatment with topoisomerase II inhibitors resemble more de
The term therapy-related myeloid neoplasms (t-MNs) is            novo AML without dysplastic features [13].
appropriate for myelodysplastic syndromes (MDS), myelo-
dysplastic/myeloproliferative neoplasms (MDS/MPN), and
acute myeloid leukemia developing secondary to cytotoxic         13.6.3 Immunophenotype
chemotherapy and/or radiation therapy. These disorders have
been combined in one entity because of their similar patho-      No specific immunophenotypic features are present in
genesis, rapid progression from t-MDS to t-AML, and their        t-MNs. Blasts often express CD34 and pan-myeloid markers
equally poor prognosis [13, 68].                                 such as CD13 and CD33. MPO may be present but can be
                                                                 downregulated. In a monocentric retrospective study of
                                                                 t-MN, immunophenotyping and immunohistochemistry
13.6.1 Epidemiology and Etiology                                showed aberrant expression of B-, T-, or NK-cell markers in
                                                                 21% and 6%, respectively [116].
An association with a prior therapy is present in up to 20% of
MDS, MDS/MPN, and AML cases [13]. The risk increases
with age. However, t-MN may also occur in the pediatric age      13.6.4 Genetics
group. Previous therapy for breast cancer is the most impor-
tant cause of t-MN followed by low-grade B non-Hodgkin           In recent years, extensive sequencing studies have identified
lymphoma [116]. Two types of t-AML occurring as a com-           recurrent mutations with diagnostic and prognostic impact.
plication of chemotherapy are recognized: first, t-MN arising    Fifteen percent of t-AML patients present with favorable
in patients treated with topoisomerase II inhibitors after a     risk fusion genes, whereas 50% have adverse cytogenetics
short period of about 1–3 years without MDS-related clinical     [117]. In t-MN developing after treatment with epipodo-
or morphological features and a better prognosis; and sec-       phyllotoxins like etoposide, fusion oncogenes like KMT2A/
ond, t-MN developing after a longer period of about 5–7          MLL-MLLT3 fusion genes are prevalent [68]. Treatment
years, for example, after therapy with alkylating agents.        with alkylating agents has been associated with adverse
These cases with a longer latency generally develop an initial   cytogenetic abnormalities such as −5/del(5q), 7/del(7q), or a
MDS phase and have a poor prognosis [68]. Other types of         complex karyotype [68]. The most frequent molecular aber-
therapy with an increased risk of a t-MN include immuno-         ration in t-AML and t-MDS affects TP53 (33%) and may
suppressive regimens for non-neoplastic disorders such as        result from the selection of a preexisting treatment-resistant
autoimmune disorders.                                            hematopoietic stem cell clone with TP53 mutation [117].
                                                                 Strong p53 immunostaining in ≥1% of bone marrow cells is
                                                                 highly predictive of a TP53 gene mutation and associated
13.6.2 Morphology                                               with adverse karyotype and poor outcome [118]. According
                                                                 to the results of mutational profiling comparing t-MN with
Bone marrow smears and core biopsies of patients with a          de novo MNs, TP53 was the most common mutated gene in
prior history of alkylating agent therapy generally exhibit      t-MDS (35.7%) as well as t-AML (33.3%), significantly
pronounced dysplastic features of all three myeloid lineages:    higher than de novo MDS (17.7%) (p = 0.0410) and de novo
a hyperplastic macro-/megaloblastic erythropoiesis with or       AML (12.8%) (p = 0.0020). t-AML showed more frequent
without ring sideroblast, numerous micromegakaryocytes,          PTPN11 but less NPM1 and FLT3 mutations than de novo
hypogranulated immature myeloid cells, and various               AML [119].
13       Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies                                                 443
Case History: Case 1 (Fig. 13.22)                                        pseudo-Pelger cells. The bone marrow contained 11%
A 75-year-old woman was admitted in a reduced general                    CD117-positive blasts with partial co-expression of CD34
condition. She reported slowly progressive weakness and                  (80%) and MPO. The karyotype was complex involving
dyspnea during the last few months. Six years earlier,                   aberrations of chromosomes 3, 5, 10, 14, 17, 21. Of note,
hysterectomy had been performed followed by local radia-                monosomy 5, del17p13, and del4q24 were detected.
 tion therapy (45 Gy GD) for endometrial adenocarcinoma.                 Molecular analysis revealed mutation of the TP53 gene. The
 The CBC was 0.5 × 109/L, Hb 6.6 g/dL, platelets                         bone marrow biopsy showed a decreased cellularity, argyro-
27 × 109/L. The peripheral blood contained 3% CD34- and                  philic fibrosis grade 2, and scattered foci of hematopoietic
 CD117-positive blasts, 6% CD19- and CD20-positive                       cells with multilineage dysplasia and excess blasts of about
 B-cells, 51% CD3- positive T-cells with co-expression of               15% (Fig. 13.21). The diagnosis was therapy-related myeloid
 CD2, CD5, CD7, and a normal T4/T8 ratio, 4% monocytes,                  neoplasm (t-MN) consistent with therapy-related MDS
 and 36% neutrophils with dysplastic features including                  (t-MDS) associated with fibrosis grade 2.
a b
MGG MGG
c d
MGG MGG
Fig. 13.22 Therapy-related myeloid neoplasm, case 1. (a–d, MGG)          cytoplasm that were also observed in the bone marrow core biopsy (e,
Bone marrow aspirate smears were hypocellular with some more cel-        f, Giemsa). Irregular reticulin fibrosis was assigned to a grade 2 (g, h,
lular areas showing a prominent dysplastic erythropoiesis with some      Gomori)
vacuolated proerythroblasts and nests of myeloblasts with a basophilic
444                      13   Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies
e f
                                                                                                    Giemsa
                                      Giemsa
g h
g h
Gomori Gomori
Case History: Case 2 (Fig. 13.23)                                           was: WBC 23 × 109/L, HB 9.3 g/dL, platelets 18 × 109/L. The
A 53-year old woman AML t(9;11)(p21.3;q23.3); KMT2A-                       peripheral blood contained 43% blasts with basophilic cyto-
MLLT3, therapy related                                                      plasm, prominent nucleoli. Blasts were positive for CD33
   A 52-year old woman was admitted with leukocytosis and                   with co-expression of MPO, CD13 and partial co-expression
thrombocytopenia. Fifteen months earlier, she was diagnosed                 of CD117 and CD14. Blasts represented 80% of nucleated
with breast cancer, NOS, grade 3, and underwent neoadju-                    cells in the bone marrow aspirate and core biopsy. Genetic
vant chemotherapy with an etoposide containing chemother-                   studies revealed a t(9;11)(p22;q23); KMT2A-MLLTT3 trans-
apy. The suspected diagnosis of her physician was bone                      location. This translocation had apparently occurred second-
marrow involvement by metastatic breast cancer complicated                  ary to cytotoxic chemotherapy. Therefore, the AML was
by a reactive leukocytosis and thrombocytopenia. The CBC                    assigned to the t-MN category and diagnosed as t-AML.
a b
HE HE
c d
NASDCL CD33
Fig. 13.23 Therapy-related myeloid neoplasm with t(9;11)                    partially NASDCL-positive (c), express CD33 (d), myeloperoxidase
(p21.3;q23.3); KMT2A-MLLT3. The bone marrow core biopsy shows a             (e), partially CD14 (f), and rarely CD34 (g). Some dysplastic mega-
dense infiltration by blasts with prominent nucleoli (a, b, H&E) that are   karyocytes were observed (h, CD61)
446                      13   Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies
e f
MPO CD14
g h
                                        CD34
                                        C
                                        CD
                                         D34
                                          34                                                             CD61
                                                                                                         CD
                                                                                                         CD6
                                                                                                           61
                                                                                                            1
13.7     AML, Not Otherwise Specified                            mutated genes per cytomorphological subtype were
                                                                  RUNX1 in M0 (43%), NPM1 in M1 (42%), DNMT3A in M2
Myeloid neoplasms can only be designated as AML, NOS              (26%), NPM1 in M4 (57%), M5a (49%), and M5b (70%),
when cytogenetic and molecular aberrations defining other         and TP53 in M6 (36%). FLT3, NPM1, and WT1 mutations
AML categories have been excluded [14]. Especially, recur-        were associated with an immature phenotype in myeloblastic
rent cytogenetic abnormalities, AML-defining mutations,           AML, whereas other combinations involving ASXL1,
multilineage dysplasia, a prior MDS or MDS-defining cyto-         RUNX1, MLL-PTD, CEBPA, or KRAS were more frequent in
genetic abnormalities, a history of cytotoxic or radiation        myeloblastic AML with maturation. Within the NPM1-
therapy, and familial predisposition have to be excluded.         mutated subcohort, ASXL1 mutations were significantly
With the exception of pure erythroid leukemia (PEL), blasts       associated with a monoblastic differentiation and DNMT3A
must represent ≥20% of blood or bone marrow cells. Within         mutations with a monocytic phenotype [123]. It is worth not-
this NOS category, subtypes are classified according to the       ing that some AML cases that are assigned to the NOS cate-
morphologic and phenotypic features of the predominant            gory at initial presentation are switched to a WHO-defined
myeloid lineage, the degree of maturation or even the clini-      cytogenetic or molecular subtype when the results of further
cal course and the association with the bone marrow archi-        analyses are available (see case 13.2.10.1).
tecture such as in acute panmyelosis with fibrosis (APMF)
[120]. Principally, the diagnostic criteria proposed by the
French-American-British (FAB) classification for AML are        13.7.1 AML with Minimal Differentiation
applied [121]. In most of the different subgroups, the AML,
NOS categories are not linked to patient prognosis. However,      In AML with minimal differentiation (AML M0 according to
the FAB subtypes still retain a current prognostic role in pre-   the FAB classification), the bone marrow comprises ≥20%
dicting the outcome of AML patients undergoing allogeneic         blasts with a small rim of cytoplasm devoid of granules or
stem cell transplantation. In a large retrospective analysis,     Auer rods [124]. Cytological or cytochemical features of a
M6/M7 patients had decreased leukemia-free survival and           myeloid differentiation such as MPO or ANAE positivity are
increased nonrelapse mortality rates [122]. The AML               absent or may be detected in <3% of blasts. However, a
phenotype- genotype associations were studied in a large         myeloid differentiation is detected by flow cytometry and/or
cohort of patients diagnosed according to FAB/WHO defini-         IH. CD34, CD117, CD13, and CD33 are often expressed.
tions by molecular profiling [123]. The most frequently           Moreover, TdT, CD7, and CD19 may be expressed.
448                                        13   Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies
Case History (Figs. 13.24 and 13.25)                                  MPO and lymphocytic markers. The karyotype was normal.
AML with morphologic and immunophenotypic features                    Initially, mutational studies were performed by a limited
suggestive of minimal differentiation. A 74-year-old man              panel. No NPM1, FLT3-ITD or TKD, CEBPA, DNMT3A,
presented with pancytopenia. The CBC was: WBC                         IDH1, or IDH2 mutations were detected. The initial diagno-
3.2 × 109/L, HB 7.6 g/dL, platelets 34 × 109/L. The periph-           sis was AML with minimal differentiation. However, retro-
eral blood contained 26% blasts with small rim of agranular           spective analysis of frozen material revealed a RUNX1
cytoplasm resembling lymphoblasts. The bone marrow aspi-              mutation and a co-operating mutation in the SRSF2 gene.
rate smears and the core biopsy were hypocellular and                 RUNX mutations are frequently detected in AML with mini-
showed the same small blast population accounting for about           mal differentiation [123]. Therefore, the case was moved to
85% of nucleated cells. Cytochemical and immunohisto-                 the provisional category AML with mutated RUNX1 (see
chemical stains revealed that only <3% of blasts contained            also Sect. 13.3.3). This example highlights that in some cases
rare MPO-positive granules. By flow cytometry, the blasts             the morphologic and immunophenotypic classification as a
were positive for CD34, HLA-DR, CD13, CD117, and TdT                  subtype of AML, NOS has to be changed later on to a more
but were negative for CD11b, CD14, CD64, CD15, and                    specifically defined AML category.
a b
                                                            MG
                                                             GG                                                                    MGG
 c                                                                    d
                                                            MG
                                                            M GG                                                                   MGG
Fig. 13.24 AML with phenotypic features of minimal differentiation.   Small-sized blasts represented about 85% of nucleated cells. (c)
(a, MGG) Blood smears showed rare blasts resembling lymphoblasts.     Scattered proerythroblasts were binucleated. (e–h) Rare blasts repre-
(b–d, MGG) The bone marrow aspirate smears were hypocellular.         senting <3% contained some MPO-positive cytoplasmic granules
13       Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies                                                449
e f
MPO MPO
g h
                                                               MPO
                                                               MP
                                                               M PO                                                                     MPO
                                                                                                                                        MP
                                                                                                                                        M P
                                                                                                                                          POO
a b
Giemsa Giemsa
Fig. 13.25 AML with phenotypic features of minimal differentiation.      TdT (e, f). MPO immunohistochemistry revealed some residual garnu-
The bone marrow core biopsy was also infiltrated by small-sized blasts   lopoietic cells and was faintly positive in <3% of the blast population
without maturation (a–c Giemsa) that strongly expressed CD34 (d) and     (g, h)
450                                   13   Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies
c d
Giemsa CD34
e f
TdT TdT
g h
                                                      MP
                                                      MPO
                                                      MPO                                                              MP
                                                                                                                       MPO
                                                                                                                       M PO
13.7.2 AML Without Maturation                                 or may even be absent, and small Auer rods may rarely be
                                                               seen. Blasts express MPO and myeloid antigens such as
AML without maturation (AML M1 according to the FAB            CD13, CD33, and CD117. CD34 and HLA-DR are found in
classification) occurs predominantly in adults with a median   about 70% of cases. Granulocytic markers (CD15, Cd65)
age of 45 years [14]. Myeloid blasts account for ≥20% of       and monocytic markers (CD14, CD64) are generally absent.
bone marrow cells. Their cytoplasmic granulation is sparse     CD7 positivity is reported in about 30% [14].
13   Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies                                               451
Case History: AML Without Maturation (Fig. 13.26)                       marrow aspirate smears and core biopsy contained the same
The patient was a 48-year-old man who was admitted to the               neoplastic population that is positive for MPO (Fig. 13.26).
hospital because of a reduced general condition and upper               The immunophenotype of blasts was as follows: positive
respiratory tract infection. The CBC was: WBC 8.3 × 109/L,              (>90%): CD117, CD13, CD33, CD38, and MPO. The karyo-
HB 6.5 g/dL, platelets 48 × 109/L. The peripheral blood smear           type was normal. Molecular analysis revealed an IDH2 muta-
showed 71% myeloblasts often with azurophilic cytoplasmic               tion that is not specific for a WHO-defined AML category.
granulation, 1% monocytes, and 28% lymphocytes. Bone                    Therefore, the diagnosis was AML without maturation.
a b
MGG MPO
c d
Giemsa Giemsa
Fig. 13.26 AML without maturation. (a, MGG) The peripheral blood        biopsy was nearly normocellular (c, d, Giemsa) and contained blasts
showed blasts with azurophilic granulation. (b, MPO) Bone marrow        that uniformly expressed CD117 (e), CD33 (f), MPO (g) but with the
aspirate smears were strongly MPO-positive and showed no maturation     exception of <2% of cells were negative for NASDCL (h)
to a more mature stage of differentiation. (c–h) The bone marrow core
452                                    13   Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies
e f
CD117 CD33
g h
MPO NASDCL
13.7.3 AML with Maturation                                     granulation typically seen in AML with inv(16)(p13.1q22)
                                                                or t(16;16)(p13.1q22). Blasts express myeloid- associated
This entity corresponds to the M2 FAB subtype and develops      antigens such as CD13, CD15, CD33, MPO, and CD117 but
predominantly in either young (<25 years) or elderly (≥60       no monocyte markers such as CD14 or CD64. CD34 positiv-
years) patients [14]. The bone marrow contains ≥20% blasts      ity may be present in some blasts. CD7 co-expression is
with a POX-positive granulated cytoplasm. A maturation to       reported in about 20–30%. There are no characteristic cyto-
promyelocytes, myelocytes, and mature neutrophils is pres-      genetic or molecular aberrations [14]. However, ASXL1,
ent in ≥10% of cells. Dysplastic features are often observed    RUNX1, MLL-PTD, CEBPA, or KRAS was more frequently
but involve <50% of cells. Basophils, mast cells, and eosino-   found in myeloblastic AML with maturation than in other
phils may be increased. These eosinophils lack the abnormal     AML, NOS subtypes [123].
13    Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies                                                  453
Case History (Fig. 13.27)                                                 58% blasts with a granulated cytoplasm, and 29% lympho-
A 67-year-old woman was diagnosed with anemia, thrombo-                   cytes. The neoplastic cells strongly expressed MP0 and were
cytopenia, and leukocytosis. The CBC was: WBC                             positive for NASDCL. No chromosomal abnormalities or
12.8 × 109/L, HB 7.3 g/dL, platelets 73 × 109/L. The periph-              mutations in BCR-ABL, CSF3R, SETBP1, ETNK1, JAK2
eral blood smear showed 3% neutrophils with pseudo-                      V617F, PDGFRA, PDGFRB, or PCM1-JAK2 genes were
Pelger-Huet anomalies, 4% metamyelocytes, 6% myelocytes,                 detected. The diagnosis was AML with maturation.
a b
MGG MPO
c d
Giemsa Giemsa
e f
NASDCL CD33
Fig. 13.27 AML with maturation. Peripheral blood smears (a, MGG;          and rarely expressed CD34 (h). Erythropoiesis and megakaryopoiesis
b, MPO) and bone marrow core biopsy (c–h) showed abundant myeloid         showed some dysplastic features that did involve less then 50% of the
cells with maturation to promyelocytes and beyond. The neoplastic         residual cells of these lineages. (a, MGG; b, MPO) Note the presence
cells were strongly positive for myeloperoxidase (b, g), NASDCL (e, f),   of some dysplastic neutrophils in (a)
454                                    13   Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies
g h
MPO CD34
13.7.4 Acute Myelomonocytic Leukemia                            populations. In AMML, mature monocytes may predomi-
                                                                 nate in the peripheral blood when compared to the bone
In acute myelomonocytic leukemia (AMML or AML M4                 marrow. Therefore, the differential diagnosis may include
according to the FAB classification), the bone marrow is         chronic myelomonocytic leukemia (CMML). However, a
infiltrated by blasts and precursors with features of granulo-   key feature of AMML is the increased number of monoblasts
cytic and monocytic differentiation. Myeloblasts, mono-          and promonocytes in the bone marrow. This immature popu-
blasts, and promonocytes must represent at least 20% of          lation is characterized by round or slightly folded nuclei
bone marrow cells. Antigens indicating a monocytic differ-       with a fine chromatin structure, small nucleoli, and a baso-
entiation such as CD14, CD64, CD11b, CD11c, CD4, CD36,           philic cytoplasm. Nonspecific cytogenetic abnormalities
CD68 (antibody PGM1), CD163, and non-specific esterase           including trisomy 8 may be present. Genetic analyses have
(ANAE) positivity are present in addition to markers of the      pointed to an association between AML with monocytic lin-
granulocytic series CD13, CD33, CD65, and CD15. On sec-          eage involvement and NPM1 mutation that was detected in
tions cut from bone marrow core biopsies, CD4, CD14, and         M4 (57%), M5a (49%), and M5b (70%) [123]. However,
CD68 positivity may be revealed by IH. There is usually a        NPM1-mutated cases have to be switched from the AML,
mixture of NASDCL-positive and -negative neoplastic sub-         NOS to the distinct category AML with mutated NPM1.
13   Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies                                               455
Case History: Case 1 (Fig. 13.28)                                    Auer rods. Monocytic cells were positive for alpha-naphthyl-
A 65-year-old man developed gingival swelling and con-               acetate-esterase (ANAE). The bone marrow core biopsy was
sulted his dentist who sent him to a hematologist. The CBC           highly hypercellular and densely infiltrated by myeloblasts,
was: WBC 15.2 × 109/L, HB 9.9 g/dL, platelets                        monoblasts, and promonocytes partially positive for CD68,
112 × 109/L. The peripheral blood smear showed 12% neu-              positive for CD33 but nearly completely negative for CD14.
trophils, 47% monocytes, 12% promonocytes, 8% mono-                  No characteristic cytogenetic or molecular aberrations were
blasts, 10% myeloblasts, and 11% lymphocytes. MPO                    found. The diagnosis was acute myelomonocytic leukemia
reaction revealed partial positivity with presence of small          (AML M4 according to the FAB classification).
a b
MGG MGG
c d
MPO ANAE
Fig. 13.28 Acute myelomonocytic leukemia case 1. (a–d) Peripheral    Dense infiltration of the bone marrow core biopsy by myelomonocytic
blood smears contained a spectrum of myelomonocytic cells (a, b,     cells (e, Giemsa) partially expressing CD68 (f), strongly positive for
MGG) some of which contained small MPO-positive Auer rods (c) or     CD33 (g) but rarely for CD34 (h)
were positive for alpha-naphthyl-acetate-esterase (ANAE, d). (e–h)
456                      13   Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies
e f
                                      Giemsa
                                                                                                          CD68
g h
CD33 CD34
Case History: Case 2 (Fig. 13.29)                                            and blasts and 26% monocytes. The neoplastic cells were
A 75-year-old woman presented with leukocytosis, anemia,                     CD34-negative but expressed CD33, CD64, CD56, CD123,
and thrombocytopenia. The CBC was: WBC 21 × 109/L, HB                        and HLA-DR and partially CD14, CD163, and myeloperoxi-
8.4 g/dL, platelets 54 × 109/L. The peripheral blood smear con-              dase. TCL1 was negative. Thirty to forty percent of bone mar-
tained 7% myeloblasts, 1% promyelocytes, 6% myelocytes,                      row cells were esterase positive. The karyotype was normal.
1% metamyelocytes, 17% neutrophils, 1% eosinophils, 2%                       No mutations were detected. Therefore, the diagnosis was
basophils, 43 monocytes, 4% promonocytes, and 18% lympho-                    acute myelomonocytic leukemia (AML M4 according to the
cytes. The bone marrow aspirate contained 49% promonocytes                   FAB classification).
a b
HE HE
c d
HE MPO
Fig. 13.29 Acute myelomonocytic leukemia, case 2. (a–h) The hyper-           especially in myeloblasts. (e) CD163 is expressed in the monocytic lin-
cellular bone marrow biopsy (a, b, H&E) was diffusely infiltrated by a       eage and in macrophages. (f) Strong expression of CD123 by the myelo-
myelomonocytic population. (c, H&E) Abundant precursors of monopoi-          monocytic population. (g) Aberrant expression of CD56. (h) Positivity
esis with pale cytoplasm and folded nuclei. (d) Partial positivity for MPO   for CD14 in a minority of the monocytic/monoblastic population
458                                 13   Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies
e f
CD163 CD123
g h
CD56 CD14
13.7.5 A
        cute Monoblastic and Monocytic                      tive for CD34 and express HLA-DR, C13, CD33, CD14,
       Leukemia                                              CD4, CD11b and/or c, CD64, CD68, CD163, and lyso-
                                                             zyme. CD7 and CD56 may be co-expressed. The cytoplasm
In both subtypes of AML, NOS (AML M5 according to            is basophilic and may contain rare azurophilic granules and
FAB classification), the monocytic lineage represents        vacuoles. The AML is designated as monocytic leukemia
≥80% of marrow cells. The percentage of blasts and pro-      when a maturation is evident and monoblasts are <80%. In
monocytes is ≥20. The neoplastic cells are positive for      acute monoblastic leukemia, the more immature mono-
alpha-naphthyl-acetate-esterase (ANAE), frequently nega-    blasts are ≥80%.
13   Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies                                                      459
Case History (Fig. 13.30)                                                 necrosis adjacent to areas diffusely infiltrated by blasts
A 53-year-old man developed abdominal pain. He was                        with gray-blue cytoplasm that were completely NASDCL
admitted to the hospital where laboratory data showed pan-                negative. By immunohistochemistry, blasts expressed
cytopenia and increased LDH (940 U/L). An abdominal                       CD33 and partially CD14 and CD163 but were negative for
mass was detected (3.3 × 3.8 × 3.7 cm). Burkitt lymphoma                  MPO. Shadows of blasts were detected within the necrosis
was suspected. The CBC was: WBC 1.3 × 109/L, HB 6.3 g/                    consistent with spontaneous tumor necrosis. Karyotyping
dL, platelets 30 × 109/L. The peripheral blood smear con-                 revealed a trisomy 8. By molecular genetic analysis, no
tained rare large blasts with round nuclei consistent with                BCR-ABL, FLT3-ITD, FLT3-TKD, NPM1, IDH1, and
monoblasts that were strongly positive for nonspecific                    IDH2 mutations were detected. The diagnosis was acute
esterase. No bone marrow aspirate could be obtained                       monoblastic leukemia (AML M5a according to the FAB
because of dry tap. The core biopsy revealed focal marrow                 classification).
a b
MGG ANAE
c d
MPO Giemsa
Fig. 13.30 Acute monoblastic leukemia. (a–c) Peripheral blood             focal areas of necrosis (d, Giemsa; f, NASDCL) adjacent to sheets of
smears. (a, MGG) Large blasts with round nuclei and blue-gray agranu-     large blasts with pale blue cytoplasm (e, Giemsa) that were negative for
lar cytoplasm. (b) Strong positivity for nonspecific esterase (b, ANAE)   NASDCL (f). Strong expression of CD33 (g) and (h) partial positivity
but (c) negativity for MPO. (d–h) The bone marrow core biopsy with        for CD14
460                      13   Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies
 e                                                f
                                                      h
Giemsa NASDCL
g h
CD33 CD14
13.7.6 Pure Erythroleukemia                                      cells with ≥30% proerythroblasts. The chromatin is dark.
                                                                  The cytoplasm of proerythroblasts is basophilic and fre-
The actual revised 2016 WHO classification restricts the          quently contains vacuoles and PAS-positive inclusions.
diagnosis of erythroleukemia (AEL) to cases characterized         There may be some megakaryocyte dysplasia in the back-
by a proliferation of immature cells of the erythroid lineage     ground [14].
constituting >80% of bone marrow cells with ≥30% pro-
erythroblasts. This rare AML subtype has been designated as       13.7.6.3 Immunophenotype
pure erythroid leukemia (PEL; [14, 125]). The former 2008         There are no immunophenotypic markers to discriminate
WHO classification listed another type of AEL: the mixed          neoplastic from non-neoplastic erythroid proliferations. The
erythroid/myeloid subtype defined by erythroid precursors         erythroid lineage expresses glycophorin C, hemoglobin,
constituting ≥50% of bone marrow cells and the presence of        CD71, and E-cadherin and may also be positive for CD117
blasts ≥20% in the non-erythroid compartment. However,            but is CD34-, MPO, and HLA-DR-negative. In poorly
studies of large patient cohorts suggested that AEL, ery-         differentiated cases, glycophorin C may be absent [14].
throid/myeloid subtype with <20% blasts may be more               CD41 and CD61 expressions have been described in some
appropriately included in the spectrum of myelodysplastic         cases leading to the differential diagnosis of acute mega-
syndromes with excess blasts rather than in AML, NOS              karyoblastic leukemia.
[126, 127]. Therefore, the mixed AEL is now split into (a)           The immunohistochemical detection of nuclear p53 over-
the categories MDS with erythroid predominance when               expression correlated to TP53 aberrations is helpful to distin-
blasts are <20% or (b) in AML with myelodysplasia-related         guish PEL from non-neoplastic erythroid proliferations and
changes (AML-MRC) when blasts are ≥20% of bone mar-               should be included in the diagnostic work-up of suspected
row cells. The negative prognostic impact of erythroid pre-       cases [130].
dominance, defined as 50% or more bone marrow erythroid
cells, in MDS is still challenging [125]. A retrospective eval-   13.7.6.4 Genetics
uation of cases previously diagnosed as AEL reclassified          Until now, no specific mutational spectrum or chromosome
slightly over half of them as MDS EB-1 or -2 according to         abnormalities have been described for PEL.
the 2017 WHO criteria. The overall survival was not differ-          The cases generally have a complex, often highly com-
ent from other patients with MDS-EB [128]. Results of             plex, karyotype [130]. Loss of chromosomes 5 and 7 or
another study suggested that cytogenetic, rather than MDS/        del(5q) or del(7q) has been reported [14].
AML subtypes, may have the most important impact on
prognosis for previously diagnosed AEL patients [129].            13.7.6.5 Caution
                                                                  • The differential diagnosis between PEL, MDS with ery-
13.7.6.1 Epidemiology                                              throid predominance, and non-neoplastic hyperplasia of
Pure erythroid leukemia is an extremely rare AML category           the erythroid lineage is challenging. Morphologic and
predominantly reported in elderly patients with a male pre-         immunophenotypic features are overlapping.
dominance. The prognosis is dismal [130].                         • Non-neoplastic erythroid-predominant conditions include
                                                                    for example megaloblastic anemia or severe hemolysis.
13.7.6.2 Morphology                                                Therefore, a significant degree of clinical, laboratory,
The erythroid lineage shows striking proliferation of               immunophenotypic, and genetic investigation are recom-
immature cells accounting for >80% of bone marrow                   mended [130].
462                                          13   Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies
Case History 1: Pure Erythroid Leukemia (PEL)                             series was strikingly reduced. About 6% of nucleated bone
(Fig. 13.31)                                                              marrow cells expressed CD34. Nearly normal numbers of
A 69-year-old male presented with anemia and leukopenia.                  megakaryocytes were present. The p53 protein was strongly
CBC: WBC 2.1 × 109/L, hemoglobin 7.5 g/dL, platelet count                 overexpressed in the majority of bone marrow cells sugges-
of 173 × 109/L. Differential count: neutrophils 61%, lympho-              tive of a TP53 mutation. Genetic studies demonstrated a
cytes 27%, monocytes 5%, eosinophils 2%, erythroblasts                    mixed lineage leukemia gene-partial tandem duplication
5%. The bone marrow biopsy was highly hypercellular with                  (MLL-PTD). No NPM1 or FLT3-ITD or -PTD mutations
a predominance of immature CD71-positive and glycopho-                    were detected. The karyotype was complex.
rin C-positive erythroblasts accounting for >85% of                          The diagnosis was acute erythroid leukemia (AEL) or pure
nucleated bone marrow cells. The MPO-positive myeloid                    erythroid leukemia (PEL) according to 2016 WHO criteria.
a b
HE HE
c d
HE CD71
Fig. 13.31 Pure erythoid leukemia (case 1). (a–c) Dense infiltration of   MPO+ myeloid cells accounting for <10% of nucleated cells. (g) High
hypercellular bone marrow spaces by blast cells (H&E). (c) Immature       KI67-positive proliferation fraction. (h) Striking nuclear overexpres-
erythroid precursors forming dense sheets (PAS). (d, e) More than 85%     sion of the p53 protein by the large majority of blasts (immunostain
of blasts belong to the erythroid lineage expressing CD71. (f) Rare       with anti-p53)
13   Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies    463
e f
CD71 MPO
g h
Ki67 p53
Case History 2: Pure Erythoid Leukemia (PEL)                          bone marrow core biopsy revealed large confluent sheets of
(Figs. 13.32 and 13.33)                                               proerythroblasts without further maturation representing >90%
A 59-year-old man was admitted because of rapidly developing          of nucleated cells. A striking feature was a strong nuclear over-
pancytopenia. The CBC was: WBC 2.2 × 109/L, HB 6.4 g/dL,              expression of the p53 protein in nearly all erythroid precursors.
platelets 24 × 109/L. Examination of bone marrow aspirate             The erythroid lineage was positive for glycophorin C, CD71,
smears revealed a striking expansion of the erythroid lineage         E-cadherin, partially CD117, and negative for CD34. Rare
accounting for >90% of cells. The cytoplasm of proerythro-            MPO-positive residual granulopoietic cells and some small
blasts was often vacuolated with PAS-positive inclusions. The         CD61-positive megakaryocytes were detected.
a b
MGG MGG
c d
MGG PAS
Fig. 13.32 Pure erythroid leukemia (case 2). (a–d) Cytology of bone   inclusions. (e–h) Trephine bone marrow biopsy showing a striking pre-
marrow aspirate smears. Abundant proerythroblasts with basophilic     dominance of the erythroid lineage (e, f, PAS) highlighted by (g, h)
often vacuolated cytoplasm (a–c, MGG) and (d) coarse PAS-positive     glycophorin C expression
13   Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies                                                  465
e f
PAS PAS
g h
Glycophorin C Glycophorin C
a b
HE CD117
Fig. 13.33 Pure erythroid leukemia (case 2). Additional immunolabel-      myeloid cells (g) accounted for about 5% and CD34-positive cells (h)
ing showed that sheets of proerythroblasts (a, H&E) expressed partially   for <1% of bone marrow cells
CD117 (b) and E-cadherin (c, d), CD71 (e), and p53 (f). MPO-positive
466                      13   Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies
c d
e f
E-Cadherin E-Cadherin
    e                                                f
                                                 h
CD71 p53
g h
MPO CD34
13.7.7 Acute Megakaryoblastic Leukemia                          ing for 50% of nucleated bone marrow cells associated with
                                                                 relevant fibrosis in most, but not all cases.
In this type of AML, NOS, the predominant blast cell popu-
lation (≥50% of blasts) expresses differentiation markers of     13.7.7.3 Immunophenotype
the megakaryocytic lineage [14]. This diagnosis can only be      Markers indicating a megakaryocytic differentiation such as
assigned to patients in whom recurrent genetic abnormalities     CD41, CD42b, and CD61 are expressed. Moreover, linker of
or a preceding MPN has been excluded (see caution).              activated T lymphocyte (LAT), factor VIII-related antigen,
                                                                 CD31, CD36, CD4b,CD13, and CD33 may be positive.
13.7.7.1 Epidemiology
Acute megakaryoblastic leukemia is a rare AML, NOS sub-          13.7.7.4 Genetics
type that may develop both in children and in adults. It may     Until now, no typical cytogenetic or mutational aberrations
occur in male patients previously diagnosed with a mediasti-     have been detected. Isochromosome 12p has been described
nal germ cell tumor. The prognosis is generally poor.            in young male patients with a mediastinal germ cell tumor
                                                                 and acute megakaryoblastic leukemia.
13.7.7.2 Morphology
On peripheral blood and bone marrow smears, medium-             13.7.7.5 Caution
sized to large megakaryoblasts show round or irregular           • For the diagnosis of acute megakaryoblastic leukemia, NOS,
nuclei with a dense chromatin structure. Irregular cytoplas-       genetic abnormalities characteristic for other well-defined
mic margins such as cytoplasmic budding may be prominent.          AML categories have to be excluded, especially transloca-
However, morphologically undifferentiated blasts may be            tion t(1;22)(p13.3;q13.1); RBM15-MKL1 or inv(3)/t(3;3)
present. In such cells, a megakaryocytic differentiation is        (q21.3q26.2) or t(3;3)(q21.3q26.2) have to be excluded.
only detected by immunophenotyping. Overt marrow fibro-          • Predominant megakaryoblastic differentiation in MPN blast
sis is present in most cases resulting in dry tap. Therefore,      phase for example of primary myelofibrosis or polycythe-
histologic evaluation of bone marrow biopsies is most help-        mia vera can mimic acute megakaryoblastic leukemia.
ful for establishing the diagnosis of an acute megakaryocytic    • Acute megakaryoblastic leukemia developing in Down
leukemia. Biopsies show confluent sheets of predominantly          syndrome is included in the category myeloid prolifera-
small-sized megakaryocytes and megakaryoblasts account-            tion associated with Down syndrome [14, 150].
468                                        13   Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies
Case History (Fig. 13.34)                                              3 with overt collagen fibrosis. There were sheets of small
An 18-year-old young male patient was diagnosed with pan-              dysplastic immature megakaryocytes and blasts mixed with
cytopenia. The CBC was: WBC 1.8 × 109/L, hemoglobin                    more undifferentiated blast cells. The megakaryoblasts rep-
6.3 g/dL, platelet count of 17 × 109/L. There was no history           resented ≥50% of cells. Precursors of the myeloid and
of a previous neoplasm or a genetic disorder. Peripheral               erythroid lineages were rarely observed. Cytogenetic and
blood smears showed some dysplastic neutrophils and rare               molecular analysis were difficult due to the low number of
medium-sized blasts with cytoplasmic blebs. Bone marrow                peripheral blasts and dry tap. So far, no abnormalities could
aspirate could not be obtained due to dry tap. The bone mar-           be detected. The diagnosis was acute megakaryoblastic leu-
row core biopsy was characterized by irregular fibrosis grade          kemia, NOS.
a b
Giemsa HE
c d
CD61 CD61
Fig. 13.34 Acute megakaryoblastic leukemia. (a–h) Bone marrow          blasts highlighted by CD61 immunolabeling (c, d). Rare presence of
core biopsy. (a, Giemsa; b, HE) Infiltration of the marrow spaces by   granulocytic precursors (e, f, NASDCL). Focally accentuated reticulin
sheets of immature atypical small megakaryocytes and megakaryo-        and collagen fibrosis grade 3 (g, h, Gomori)
13   Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies       469
e f
NASDCL NASDCL
g h
GOM GOM
13.7.8 Acute Basophilic Leukemia                              and in some cases also PAS. They are negative for myeloper-
                                                               oxidase and nonspecific esterase.
According to 2016 WHO classification, basophilic leukemia
is a very rare myeloid neoplasm with a primary differentia-    13.7.8.2 Immunophenotype
tion into basophils [14]. Other myeloid neoplasms that may     Blasts are MPO-, CD117 negative, but positive for CD33,
contain increased numbers of basophils have to be excluded.    CD13, CD9, CD11b, CD123, and CD203c. They may be
Examples include BCR-ABL-positive CML in accelerated or        tryptase positive but in contrast to mast cells do not co-express
blast phase, or t(6,9) AML. In a consensus paper, the term     CD117. They may be positive for CD34, HLA-DR, and TdT.
“hyperbasophilia” (HB) was proposed for cases with a persis-
tent peripheral basophil count ≥1000/μL and considered to be   13.7.8.3 Genetics
highly indicative of the presence of an underlying myeloid     No typical genetic abnormalities have been described in
neoplasm [131]. No epidemiologic data are available.           acute basophilic leukemia. Male infants may carry t(X;6)
                                                               (p11.2; q23.3). The 2016 WHO classification also include
13.7.8.1 Morphology                                           cases with t(3;6) or 12p abnormalities and basophilia into
Medium-sized blast cells show a basophilic cytoplasm that      this leukemia NOS category.
contains coarse basophilic granules. However, these granules
are predominantly visible on peripheral or bone marrow         13.7.8.4 Caution
aspirate smears. Granules are highlighted by metachromatic     • Basophilia may occur in BCR-ABL-positive CML in
staining with toluidine blue. Basophilic granules may not be     accelerated or blast phase.
detected on conventionally stained blood and bone marrow       • Other well-defined myeloid neoplasms such as AML with
smears and BMTB sections. Ultrastructural analysis pro-          t(6,9) must be considered as differential diagnosis and do not
vides evidence of characteristic granules. Using cytochemis-     fit in the category of acute basophilic leukemia.
try, blasts lack naphthol-ASD-chloroacetate-esterase in        • In contrast to mast cell neoplasms, acute basophilic leu-
contrast to mast cells but are positive for acid phosphatase     kemia is negative for CD117 but may express tryptase.
13   Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies                                                                   471
Case History: Acute Basophilic Leukemia (Fig. 13.35)                             of nucleated cells with a predominance of basophils that
A 74-year-old woman presented with anemia and throm-                             were partially positive for tryptase but negative for CD117.
bocytopenia. The CBC was: WBC 5.9 × 109/L, HB 9.1 g/                             CD34 was expressed in a subset of blasts. Erythropoiesis
dL, MCV 101 fL, platelets 38 × 109/L. The peripheral                             showed macroblastic features. Megakaryocytes were
blood smears showed 38% immature basophils with coarse                           rarely present. The nuclei of the majority of blasts were
granules that also predominated in the bone marrow aspi-                         strongly labeled by a p53 antibody. No further genetic
rate smears. The core biopsy was only slightly hypercel-                         studies were performed. The diagnosis was acute baso-
lular and was infiltrated by blast cells accounting for 70%                      philic leukemia.
a b
MGG MGG
c d
MGG HE
Fig. 13.35 Acute basophilic leukemia. (a–c) Bone marrow aspirate smears          a pale basophilic cytoplasm (d, H&E; e, f, Giemsa). (f) Faint basophilic
showing numerous blasts containing coarse basophilic granules (MGG). (d,         granules detected in rare blasts at high magnification. (g) Blasts are negative
e) Slightly hypercellular bone marrow core biopsy infiltrated by medium-         for CD117 while (h) a subpopulation expresses tryptase
sized blasts with round or slightly irregular nuclei, one or two nucleoli, and
472                      13   Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies
e f
Giemsa Giemsa
g h
CD117 Tryptase
13.7.9 Acute Panmyelosis with Myelofibrosis                         marrow that may also contain aggregates of lymphoid cells.
                                                                     Fibrosis scored as grade 2 or 3 has to warrant this diagnosis.
This panmyeloid proliferation involves all three myeloid lin-
eages and is associated with bone marrow fibrosis grade 2–3 [14,     13.7.9.2 Genetics
120]. Additional criteria are a rapid onset of the disease without   Most cases exhibit cytogenetic and/or molecular abnormali-
a preceding MDS, MDS/MPN, or MPN and absence of spleno-              ties that, however, are not specific for this disorder. It has
megaly and leukoerythroblastosis. Most patients are pancytope-       been proposed that cases with a complex karyotype involv-
nic. No epidemiologic data are available since this disorder is      ing abnormalities of chromosomes 5 and/or 7 should rather
very rare. Most patients who have been reported are elderly.         be classified as AML-MRC. OncoScan copy number and
                                                                     mutational analysis have shown high prevalence of TP53
13.7.9.1 Morphology and Immunophenotype                             abnormalities and chromosomal aneuploidy in APMF [120].
Pancytopenia is reflected by peripheral blood smears. Blasts
are generally rare. No teardrop cells should be present, but rare    13.7.9.3 Caution
erythroblasts may be observed. Bone marrow blast cells that          The differential diagnosis include
often express CD34 are generally ≥20%, but their number is           • Hyperfibrotic variants of MDS-EB-2.
frequently not very high. Atypical CD42 or CD61+ megakary-           • AML-MRC in cases with MDS-related cytogenetic
ocytes are prominent but do not fulfill the criteria of acute          abnormalities.
megakaryoblastic leukemia. In addition, dysplastic erythroid         • MPNs with overt three lineage dysplasia and rapidly
and immature granulocytic cells are present in the fibrotic            developing fibrosis.
474                                           13   Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies
Case History (Fig. 13.36)                                                  revealed a diffuse reticulin fibrosis grade 2. Atypical predomi-
A 72-year-old woman was admitted because of persistent                     nantly small-sized megakaryocytes accounted for about 35%
fever. She reported to have been in good health until 8 weeks              of bone marrow cells. Erythropoiesis was dysplastic and mac-
earlier when fatigue developed. The CBC was: WBC                           roblastic. Granulopoiesis showed immature dysplastic precur-
1.6 × 109/L, Hb 6.8 g/dL, platelets 28 × 109/L. By physical                sors. CD34+ small blast cells accounted for about 30% of
examination, no lymphadenopathy or hepatosplenomegaly                      nucleated cells. CD34 was also expressed by a subset of small
was observed. The peripheral blood smears showed pancyto-                  dysplastic megakaryocytes. Moreover, T-cells were increased
penia. Small-sized blasts predominantly positive for myelo-                in the bone marrow stroma. A TP53 mutation das detected. No
peroxidase accounted for 40% of the rare nucleated cells. No               AML-defining molecular or cytogenetic abnormality was
erythroblasts or dacrocytes were present. Bone marrow aspi-                reported. The diagnosis was acute panmyelosis with
rate resulted in a dry tap. The bone marrow core biopsy                    myelofibrosis.
a b
Gomori Gomori
c d
Giemsa CD61
Fig. 13.36 Acute panmyelosis with myelofibrosis (APMF). (a, b)             small blast cells. (f) Immature NASDCL-positive cells of the granulo-
Dense reticulin fibrosis grade 2 (Gomori’s reticulin stain). (c, Giemsa)   cytic lineage adjacent to dysplastic megakaryocytes. (g, MGG) Rare
Numerous dysplastic predominantly small megakaryocytes accounting          blasts are observed on peripheral blood smears that are positive for
however for less than 50% of bone marrow cells express CD61 (d) and        MPO (h)
CD34 (e). Note also the presence of clusters formed by CD34-positive
13   Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies       475
e f
CD34 NASDCL
g h
MGG MPO
Myeloid sarcomas (MS) formerly also referred to as chlo-          MS are composed of neoplastic myeloid cells with or with-
roma are defined as extramedullary tumor-forming extra-           out maturation lacking specific morphologic or immunophe-
medullary masses of myeloid blasts with or without                notypic features [15]. They may express MPO, CD33, CD34,
maturation [15, 132, 133]. Even if these tumors occur by          and CD117 and show reactivity for naphthol-ASD-
definition outside the bone marrow, they are briefly men-         chloroacetate-esterase. However, a monoblastic or myelo-
tioned here, because their diagnosis has the same impact as       monocytic phenotype is frequently reported and characterized
the diagnosis of AML. Diffuse infiltrates of extramedullary       by the expression of CD14, CD11c, and CD163. However,
sites by myeloid blasts for example in the oral mucosa or the     morphologic and immunophenotypic equivalents to other
skin do not fulfill criteria of an MS [15].                       AML subtypes such as CD61+CD41+ acute megakaryoblas-
                                                                  tic leukemia or CD71+ glycophorin C+ hemoglobin+ ery-
                                                                  throid leukemia may also occur.
13.8.1 Epidemiology and Clinical Features
Case History: Myeloid Sarcoma (Fig. 13.37)                              performed and showed a dense infiltration by medium-
A 23-year-old male patient developed a mediastinal                      to large-sized polymorphic blasts with prominent nucle-
mass with a maximal diameter of 12 cm. The CBC was:                     oli and a basophilic cytoplasm strongly co-expressing
WBC 15 × 10 9/L, HB 14.2 g/dL, platelets 280 × 10 9/L. The              CD33 and CD43 and a high Ki67 proliferation fraction
peripheral blood smears contained 3% band forms, 68%                    >75%. About 65% of blasts expressed CD71 partially in
neutrophils, 3% eosinophils, 4% monocytes, and 22%                      co-expression with CD117. Smaller subpopulations were
lymphocytes. No dysplastic cells or blasts were observed.               positive for CD68, CD4, lysozyme, and myeloperoxi-
The bone marrow was normocellular and showed a regu-                    dase (Fig. 13.37). No genetic testing was performed. The
lar maturation of all hematopoietic lineages. A mediasti-               diagnosis was myeloid sarcoma with an isolated
noscopic wedge biopsy of the mediastinal mass was                       extramedullary manifestation.
a b
H&E H&E
c d
H&E CD33
Fig. 13.37 Myeloid sarcoma. (a–c) H&E stained sections of the medi-     population expresses CD33 (d), CD43 (e), and CD71 (f) in the majority
astinal mass showing polymorphous medium- to large-sized blasts with    of cells. (g) Focal positivity for CD117. (h) High Ki67+ proliferation
prominent nucleoli intermingled with some eosinophils. The neoplastic   fraction
478                      13   Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies
e f
CD43 CD71
g h
CD117 Ki67
Case History 1: Bone Marrow Failure and AML                               cytes. Examination of bone marrow aspirate smears showed
Associated with Germline GATA2 Mutation (Figs. 13.38                      69% CD33-positive myeloblasts co-expressing CD33 and
and 13.39)                                                                HLA-DR. Blasts were partially positive for CD7, CD117,
A 32-year-old female patient had been diagnosed with tri-                 and CD34. In addition, 2% promyelocytes, 1% eosinophils,
cytopenia and prominent monocytopenia suggestive of                       3% plasma cells, 11% lymphocytes, and 14% dysplastic
hypoplastic MDS 3 years earlier. The bone marrow core                     erythroid precursors were present, but no megakaryocytes
biopsy was moderately hypocellular and showed small dys-                  could be detected. The bone marrow core biopsy was hypo-
plastic megakaryocytes, rare erythroid islands, and clusters              cellular in relation to patient age with 65% fat cells that
of small megakaryocytes with completely separated or soli-                surrounded a prominent interstitial infiltrate by medium-
tary nuclei (Fig. 13.38). The differential diagnoses included             sized blasts with prominent nucleoli and a basophilic cyto-
toxic or drug-induced bone marrow injury or bone marrow                   plasm and a strong aberrant cytoplasmic NPM1
failure. She reported recurrent pulmonary infections.                     immunolabeling (Fig. 13.39).
Subsequently, the patient presented with cutaneous and                       Next-generation sequencing revealed a NPM1 mutation
mucosal hemorrhage. Hematologic evaluation revealed                       and a germline GATA2 mutation that was detected in DNA
pancytopenia. The CBC was: WBC 3.2 × 109/L, HB 7.1 g/                     probes extracted from both the bone marrow and the nails.
dL, platelets 12 × 109/L. The peripheral blood smears con-                   The diagnosis was NPM1-mutated AML developing in a
tained 22% myeloblasts, 3% myelocytes, and 75% lympho-                    patient with germline GATA2 mutation.
a b
MGG Giemsa
c d
                                                             Giemsa
                                                                                                                                    NASDCL
Fig. 13.38 Initial biopsy. Bone marrow failure associated with germ-      reduced granulopoiesis.(e, f) Loose clusters of megakaryocytes are
line GATA2 mutation. (a–c, Giemsa). Moderately hypocellular bone          highlighted by CD61 immunohistochemistry. (g) Interstitial and micro-
marrow spaces with rare erythroid islands, predominantly small mega-      focal CD3-positive T-cells. (h). Small nests of CD34-positive precur-
karyocytes, and (d, Naphthol-ASD-chloroacetate esterase) a left-shifted   sors accounting for less than 5% of nucleated bone marrow cells
13    Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies                                                   481
e f
CD61 CD61
g h
CD3 CD34
a b
                                                          Giemsa
                                                                                                                                    Giemsa
Fig. 13.39 Sequential biopsy. AML associated with germline GATA2      sized blast cells that are (c, d) partially positive for naphthol-ASD-chlo-
mutation. (a, b) The bone marrow core biopsy is hypocellular for a    roacetate-esterase, (e) CD117, and (f) CD7. (g, h) Note the strong
32-year-old woman and shows an interstitial infiltration by medium-   aberrant cytoplasmic positivity for NPM1
482                      13   Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies
c d
                                    NASDCL
                                                                                                        NASDCL
e f
CD117 CD7
g h
NPM1 NPM1
Case History 2: AML Associated with Germline CBL                          and CD33. In addition, 35% CD14+ monocytes were pres-
Mutation (Fig. 13.40)                                                     ent. FISH analysis showed CBFB(16q22) to MYH11
A 44-year-old woman suddenly developed ecchymoses and                     (16p13.1)-rearrangement in >60% of bone marrow cells.
gastrointestinal bleeding. The CBC was: WBC 12 × 109/L, HB                   The initial diagnosis was AML with inv(16) and a myelo-
6.3 g/dL, platelets 15 × 109/L. The differential count was as             monocytic phenotype.
follows: 41% blast cells, 2% myelocytes, 1% m etamyelocytes,                Additional genetic testing revealed a biallelic CBL
2% band forms, 12% neutrophilic granulocytes, 31% mono-                   mutation consistent with a germline CBL mutation.
cytes, and 11% lymphocytes. Rare erythroblasts were observed.             Germline CBL mutations are generally associated with
   The bone marrow core biopsy was strikingly hypercellu-                 juvenile myelomonocytic leukemia (JMML). The medical
lar and diffusely infiltrated by myeloid blasts and myelo-                history did not provide any hint to a previous hematologic
monocytic cells associated with bone marrow fibrosis grade                disorder. However, it has been reported that JMML in
1 (Fig. 13.40).                                                           patients with germline CBL mutations often regresses
   Immunophenotyping reported 54% CD34+ CD117+                            spontaneously and may not have been diagnosed in our
MPO+ bone marrow blasts partially co-expressing CD13                      patient [149].
a b
MGG
c d
Giemsa Gomori
Fig. 13.40 AML associated with germline CBL mutation. (a, b)              marrow core biopsy specimen (Giemsa) showing (d) reticulin fibrosis
Peripheral blood films containing blasts, dysplastic monocytes, neutro-   grade 1 (Gomori) and (e, f) a dense infiltration by a predominantly
phils, and rare erythroblasts (MGG). (c) Strikingly hypercellular bone    naphthol-ASD-chloroacetate-esterase-positive myeloid population
484                      13   Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies
e f
NASDCL NASDCL
13.10 M
       yeloid Proliferations Associated                            ally >10%. In addition, the diagnosis requires the detection
      with Down Syndrome                                            of a GATA1 mutation (see below). Typically, the blast cells in
                                                                    TAM have a megakaryoblastic morphology and phenotype.
Individuals with constitutional trisomy 21 (Down syn-               They show cytoplasmic blebbing and express CD42 and
drome, DS) have a significantly increased risk to develop           CD61 in co-expression with CD34, CD117, CD33, and other
different types of hematopoietic neoplasms especially               myeloid markers but are negative for myeloperoxidase
myeloid proliferations in infancy, childhood, adolescence,          CD15, CD14, or glycophorin A. An aberrant expression of
and adulthood [16].                                                 CD56 and CD7 may be present. In addition, basophilia may
                                                                    be observed.
                                                                       In cases designated as silent TAM, no or very low num-
13.10.1     Epidemiology and Clinical Findings                     bers of blasts are present despite a GATA1 mutation. These
                                                                    infants are also at risk to develop ML-DS.
Trisomy of chromosome 21 has been reported to be present               ML-DS may present with pancytopenia and MDS-like
in one in 700–1000 live births worldwide [150]. In children         features associated with low percentages of peripheral
with DS myeloid leukemia (ML-DS), and acute lymphoblas-             blasts and macrocytic erythroid cells. The bone marrow
tic leukemia are increased by 150- and ∼30-fold, respec-            shows marked dysplastic features. Overt AML is fre-
tively [151, 152]. About 10% of neonates and also some              quently associated with bone marrow fibrosis, resulting in
fetuses with DS present with neonatal hematopoietic disor-          a dry tap. Dysplastic CD42+CD61+ megakaryocytes and
der that is referred to as transient abnormal myelopoiesis          megakaryoblasts are increased, often associated with a
(TAM). TAM may be the first symptom of infants with                 megaloblastic erythropoiesis and dysgranulopoiesis. The
mosaicism for trisomy 21 without the characteristic pheno-          immunophenotype of blasts in ML-DS is similar to that
type of DS. TAM may be detected as an incidental finding on         seen in TAM.
review of a blood film in asymptomatic infants or as a conse-          AML in patients >5 years are not different from AML in
quence of clinical symptoms such as coagulopathy, multior-          non-DS patients.
gan failure and extramedullary infiltrates associated with
hepatosplenomegaly and effusions [152]. In the majority of
infants, spontaneous remission of TAM occurs. However,              13.10.3     Genetics
about 20–30% of infants develop a myeloid leukemia [152]
especially acute megakaryoblastic leukemia 1–3 years later.         Trisomy 21 is associated with a perturbation of fetal hematopoi-
AML may develop secondary to a myelodysplastic-syndrome            esis preceding the acquisition of GATA1 mutations [152, 153].
(MDS)-like phase. Both MDS and AML in children with                 The fetal liver of DS-fetuses harbors increased dysplastic mega-
Down syndrome are designated as acute leukemia associated           karyocytes while platelet counts are reduced. Next-generation
with DS (ML-DS). AML in children with Down syndrome                 sequencing-based methodology has recently shown that GATA1
younger than 5 years is 150-fold increased compared to              mutations have been detected in up to 30% of infants with DS
healthy children. Moreover, the risk to develop a myeloid           and in all patients with TAM or ML-DS [154]. GATA1 is the key
neoplasm is also significantly higher in adult patients with        hematopoietic transcription factor. TAM has been attributed to
DS than in the general population. However, a globally              the cooperation between constitutional trisomy 21 and acquired
decreased incidence of solid tumors as compared to age-            somatic N-terminal truncating mutations in GATA1. Additional
adjusted non-DS controls has been reported [150].                   genetic alterations including those in epigenetic regulators and
                                                                    signaling molecules are involved in the progression from TAM
                                                                    to ML-DS [155]. Examples are mutations of CTCF, EZH2,
13.10.2     Morphology and Immunophenotype                         KANSL1, JAK2, JAK3, MPL, SH3B3, and RAS pathway genes
                                                                    [155]. Myeloid neoplasms developing in children with DS are
It is important to realize that blasts may be seen in the periph-   similar to non-DS MDS or AML, may not harbor GATA1 muta-
eral blood smears of neonates with DS that do not develop           tions but may show trisomy 8 in nearly half of the cases while
TAM. In infants with TAM, the blast cell counts are gener-          monosomy 7 is very rare [16].
486                                          13   Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies
Case History 1: Intrauterine Transient Abnormal                           nodes and thymus were observed in addition to congenital
Myelopoiesis (TAM) (Fig. 13.41)                                           heart disease. Microscopic evaluation revealed abundant
This preterm born male infant of 35 weeks of gestational age              intravascular blasts associated with extravascular infiltrates in
was diagnosed with trisomy 21, a WBC of 120 × 109/L,                      multiple organs. Liver and spleen showed a striking intrasinu-
thrombocytopenia, liver failure, ascites, and coagulopathy.               soidal expansion of CD34+ hematopoietic precursors and
He passed away from multiorgan failure 7 h after birth. Blood             blasts predominantly of the megakaryocytic lineage express-
films showed 72% blasts, rare hypogranulated neutrophils,                 ing CD61. In addition, rare MPO+ granulocytic precursors
some circulating micromegakaryocytes, megakaryoblasts,                    and dysplastic erythroid cells were seen. Molecular analysis
and macrocytic red blood cells. At autopsy, hepatospleno-                 revealed a mutation in exon 2 of the GATA1 gene. The diag-
megaly and enlargement of multiple organs including lymph                 nosis was TAM in Down syndrome.
a b
H&E H&E
c d
H&E HE
Fig. 13.41 Down syndrome, intrauterine TAM. (a, b) Meningeal blood        some larger abnormal megakaryocytes (c, d, H&E) that predominantly
vessels stuffed with abundant blast cells (H&E). (c–g) Hepatic involve-   co-express CD61 (e, f). (g) Rare intrahepatic foci of MPO+ granulocytic
ment by TAM: The hepatic sinusoids are dilated due to immature hema-      precursors. (h) Involvement of the thymus by the myeloid proliferation
topoietic cells including megakaryoblasts, micromegakaryocytes, and       associated with a destruction of Hassal bodies (H&E)
13   Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies     487
e f
CD61 CD61
g h
MPO H&E
Case History 2: Myeloid Leukemia Associated with                             aspirate resulted in a dry tap. The bone marrow core biopsy
Down Syndrome (Fig. 13.42)                                                   revealed reticulin fibrosis grade 2 with small foci of collag-
A 2-year-old boy had a history of TAM in the neonatal                        enous fibrosis and increased content of CD3+ lymphocytes.
period that spontaneously resolved within 3 months. He                       There was a diffuse infiltration by CD34+ blasts including
subsequently developed anemia and thrombocytopenia. At                       >50% CD61+ megakaryoblasts and small atypical mega-
admission to the Pediatric Hematology-Oncology                               karyocytes. By genetic testing, a JAK2 mutation in addition
Department, the CBC was: WBC 2.9 × 109/L, HB 5.8 g/dL,                       to GATA2 mutation, a trisomy 21 and a monosomy 7 were
platelets 25 × 109/L. The differential count showed 12%                      detected.
blasts with cytoplasmic blebbing, some dysplastic neutro-                       The diagnosis was myeloid leukemia associated with
phils including pseudo-Pelger cells, lymphocytes, and occa-                  Down syndrome (ML-DS) consistent with acute megakaryo-
sionally teardrop-shaped red blood cells. Bone marrow                       blastic leukemia.
a b
Gomori Gomori
c d
Giemsa Giemsa
Fig. 13.42 Myeloid leukemia associated with Down syndrome. (a–h)             showing features of abnormal megakaryocytes (Giemsa). (e) Interstitial
Bone marrow core biopsy. (a, b) Reticulin fibrosis grade 2 with small        and focal increased inconspicuous CD3+ lymphocytes. (f) High content
foci of collagenous fibrosis (Gomori reticulin stain). (c, d) Infiltration   of CD34+ precursor/blast cells with (g, h) a predominance of CD61+
of bone marrow spaces by small- to medium-sized atypical cells often         megakaryoblasts and small atypical megakaryocytes
13   Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies      489
e f
CD3 CD34
g h
CD61 CD61
13.11 B
       lastic Plasmacytoid Dendritic Cell                       menting the diagnostic specificity alongside CD4, CD56,
      Neoplasm                                                   CD123, and TCL1 [160]. Numerous other antigens such as
                                                                 CD7, CD33, CD38, CD71, TdT, and BCL2 may be pres-
Blastic plasmacytoid dendritic cell neoplasm (BPDCN) is an       ent. A dot-like reactivity for CD68 is frequently seen.
aggressive myeloid malignancy that involves skin, bone mar-      Some cases may lack CD4 or CD56 immunoreactivity.
row, peripheral blood, lymph nodes, and spleen. This rare        BPDCN cases are negative for CD34, MPO, CD117,
WHO-defined entity is actually considered to be histogeneti-     CD14, CD163, and lysozyme.
cally related to the precursors of interferon type-1 producing
plasmacytoid dendritic cells. BPDCN was previously referred
to as CD4+CD56+ hematodermic neoplasm [156, 157].                13.11.3    Genetics
Case History (Figs. 13.43 and 13.44)                                     CD303, TCL1, TCL4, CD7, and BCL2 but negativity for
A 76-year-old male patient developed multiple reddish-                  CD34, CD14, and myeloperoxidase. The bone marrow tre-
brown maculo-papular skin lesions. Clinical examination                  phine biopsy showed a focally accentuated infiltration by the
revealed anemia, thrombocytopenia, and leukocytosis. The                 blast cells. In addition to the above-mentioned antigens, a
peripheral blood, bone marrow smears, and skin biopsy                    weak expression of CD71 and a dot-like positivity for CD68
showed medium-sized blasts with irregular nuclear contours,              were observed. The residual hematopoiesis showed dysplas-
small nuclei, and an agranular cytoplasm. Some blasts had a              tic features. The genetic profile was characterized by muta-
hand-mirror-shaped      cytoplasm.     Immunophenotyping                 tions in TET2 and NRAS. The diagnosis was BPDCN
revealed expression of CD4, CD56, CD123, CD43, BDCA-2/                   involving skin, bone marrow, and blood.
a b
MGG Giemsa
c d
Giemsa CD4
Fig. 13.43 Blastic plasmacytoid dendritic cell neoplasm (BPDCN),         granulocytic precursors. (b, c) Focally accentuated infiltration of the
bone marrow. (a) Bone marrow smear showing numerous medium-             bone marrow core biopsy by blasts with irregular nuclear contours,
sized partially hand-mirror-shaped agranular blast cells with an imma-   small nucleoli, and a grayish-blue cytoplasm. (d–h) Blast cells express
ture chromatin structure. Note the presence of some dysplastic           CD4 (d), CD123 (e, f), TCL1 (g), and CD7
492                      13   Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies
e f
CD123 CD123
g h
TCL1 CD7
a b
H&E H&E
c d
CD123 CD123
e f
CD56 CD4
Fig. 13.44 41. Blastic plasmacytoid dendritic cell neoplasm (BPDCN),       mis. (c–f) Immunohistochemical staining of the skin biopsy revealing
skin biopsy (a, b), H&E-stained sections of the skin biopsy showing        expression of CD123 (c, d), CD56 (e), and CD4 (f) by blast cells
dermal infiltration by medium-sized neoplastic cells sparing the epider-
494                                        13   Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies
13.12 A
       cute Leukemias of Ambiguous                             cells of AUL are generally small- to intermediate-sized with
      Lineage                                                   round or irregularly shaped nuclei, stippled chromatin, and
                                                                agranular cytoplasm. They may resemble blasts from AML
Ambiguous lineage acute leukemias (ALAL) represent a with minimal differentiation or acute lymphoblastic leuke-
heterogeneous group of hematopoietic neoplasms that can- mia. They do not express markers that are lineage-specific,
not clearly be assigned either to the myeloid or lymphoid especially not myeloperoxidase.
lineage through immunophenotyping [163]. The category of           MPAL may either comprise a single uniform population
ALA includes undifferentiated leukemias that lack lineage- of blasts showing a differentiation toward both lymphoid
specific antigens and mixed-phenotype acute leukemias and myeloid lineages or a hybrid morphology characterized
(MPALs; [163]). In some MPALs, blasts may simultane- by two distinct lymphoid and myeloid populations [165,
ously express markers of both the myeloid and B/T lymphoid 166]. Myeloid blasts may exhibit features of AML without
lineages, thus exhibiting a biphenotypic pattern. Other cases maturation or correspond to monoblasts. The diagnostic
of MPAL may harbor two distinct blast populations that each work-up of MPAL must include flow cytometry immuno-
fulfill criteria for B-cell, T-cell, or myeloid differentiation phenotyping and immunohistochemistry using a large
and may be designated as bilineal.                              panel of antibodies. In addition, cytochemistry studies for
                                                                myeloperoxidase and nonspecific esterase are essential
                                                                [167].
13.12.1 Epidemiology and Clinical Features                        The criteria for the assignment to lineage commitment
                                                                have been defined as follows (see Table 13.9): According to
ALALs are diagnosed predominantly in adults but may also Béné, myeloid engagement is characterized by the expres-
develop in children. These hematopoietic malignancies are sion of myeloperoxidase or, for cells of the monocytic lin-
rare and account for <4% of acute leukemias, but the exact eage, by the expression of at least two of the
incidence is unknown [163]. The so-called biphenotypic monocyte-associated antigens CD11c, CD14, CD64, lyso-
subtype of MPAL is considered to be more frequent than the zyme, and/or nonspecific esterase activity demonstrated in
bilineal subtype. ALALs are poor prognosis leukemias [164]. cytochemistry. T-lineage commitment is characterized by
                                                                cytoplasmic or surface (rare) expression of the epsilon chain
                                                                of CD3. B-lineage engagement is characterized by strong
13.12.2 Morphologic                                            CD19 expression associated with at least one of the follow-
              and Immunophenotypic Features                     ing: cytoplasmic or membrane expression of CD79a, CD22,
                                                                and/or CD10 surface labeling. If CD19 is weakly expressed,
According to the revised 2016 WHO classification, six dif- two of the latter markers must be present to confirm B-lineage
ferent entities are designated as ALALs (Table 13.8). Blast features [2, 165].
Table 13.9 Criteria for lineage assignment for a diagnosis of MPAL         the regulation of either epigenome or transcription such as
Lineage assignment criteria                                                DNMT3A, RUNX1, TP53, and KMT2D (MLL2) and in
Myeloid lineage                                                            genes involved in DNA repair pathway [168]. In addition,
 Myeloperoxidase (flow cytometry, immunohistochemistry, or                numerous nonspecific other mutations have been detected
   cytochemistry)
                                                                           in ALAS.
 Monocytic differentiation (at least two of the following:
   nonspecific esterase cytochemistry, CD11c, CD14, CD64,
   lysozyme)
T-lineage                                                                  13.12.4     Caution
 Stronga cytoplasmic CD3 (with antibodies to CD3 ε chain)
 Surface CD3                                                              • Acute leukemias with genetic features of other well-
B-lineage
                                                                             defined WHO entities are excluded from the ALAL
 Stronga CD19 with at least one of the following strongly
   expressed: CD79a, cytoplasmic CD22, or CD10                               category.
Or                                                                         • Examples include leukemias with t(8;21), inv16, or
Weak CD19 with at least two of the following strongly expressed:             t(15;17) or myeloid neoplasms with FGFR1 rear-
CD79a, cytoplasmic CD22, or CD10                                             rangement even if they exhibit a bilineage
Adopted from Arber et al. [2]                                                differentiation.
a
 Strong defined as equal or brighter than the normal B or T cells in the   • Chronic myeloid leukemia in blast crisis, AML with
sample
                                                                             myelodysplasia-related changes, and therapy-related
                                                                             AML also do not fit into this category.
13.12.3       Genetics                                                    • Various AML cases may harbor aberrant expression of
                                                                             lymphoid-lineage-associated antigens such as CD7 but do
The genetic findings in ALALs are heterogeneous (see                         not fulfill the criteria listed in Table 13.9 that are required
Table 13.9). By whole-exome sequencing and transcrip-                        for the diagnosis of ALAL.
tome sequencing, common gene mutations seen in hema-                       • Acute B- or T-lymphoblastic leukemia may aberrantly
topoietic neoplasms were found in the majority of ALAL                       express myeloid-associated antigens such as CD13,
cases [168]. Most mutations occurred in genes involved in                    CD33, and/or CD117.
496                                        13   Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies
Case History. Bilineal MPAL B/Myeloid, NOS                             cytometry confirmed the presence of two distinct population:
(Figs. 13.45 and 13.46)                                                The lymphoblastic population was CD19, CD10, partially
A 74-year-old male patient was admitted with the diagnosis             CD20, and TdT positive while the myeloid population
of acute leukemia. The CBC was: WBC 35 × 109/L, HB                     expressed myeloperoxidase, CD33, and partially CD14 and
9.3 g/dL, platelets 25 × 109/L. Physical examination revealed          CD15. The bone marrow core biopsy was diffusely infil-
a mild hepatosplenomegaly. On cytological smears, the                  trated by blasts that also exhibited a dimorphic phenotype.
peripheral blood and bone marrow aspirate showed a dimor-              The majority of blasts expressed CD19, CD10, PAX5, and
phic blast cell population: about 65% of blasts were small             TdT while a minority of immature cells were positive for
and resembled lymphoblasts and were myeloperoxidase neg-               myeloperoxidase, CD33, and partially Cd15 and CD14. No
ative while about 35% of blasts were of larger size with more          further genetic analyses were performed. The diagnosis was
abundant cytoplasm and expressed myeloperoxidase. Flow                 bilineal MPAL B/myeloid, NOS.
a b
MGG MGG
c d
MGG MGG
Fig. 13.45 Bilineal MPAL B/myeloid, NOS. (a, b) Peripheral blood       biopsy is densely infiltrated by blasts (Giemsa) that are positive for
smears and (c, d) bone marrow aspirate smears showing a dimorphic      CD34 (f). About 35% of blasts show features of immature myeloid cells
blast cell population comprising small lymphoblasts and larger         that express myeloperoxidase (g, h)
myeloblast-like cells (MGG). (e) The hypercellular bone marrow core
13   Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies                                               497
e f
Giemsa CD34
g h
MPO MPO
a b
CD15 CD14
Fig. 13.46 Bilineal MPAL B/myeloid, NOS. (a, b) Myeloid blasts are partially positive for CD15 (a) and CD14 (b) and express CD33 (c). (d–h)
The majority of blasts belongs to the B-cell lineage expressing CD19 (d, e), partially CD20 (f), strongly PAX5 (g), and TdT (h)
498                      13   Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies
c d
CD33 CD19
e f
CD19 CD20
g h
PAX5 TdT
13.13 P
       recursor Lymphoid Neoplasms:                                Table 13.10 Subgroups of T-ALL according to antigen expression
      T-Lymphoblastic Leukemia/Lymphoma                             Subgroup       Antigen expression
                                                                    Pro-T (T-I)    cCD3+/CD7+/CD2−/CD1a/CD34+/−;
T-cell acute lymphoblastic leukemia/lymphoma (T-ALL/                Pre-T (T-II)   cCD3+/CD7+/CD2+/CD5+/−CD1a−/CD34+/−;
LBL) is an aggressive malignancy derived from T-cell pro-           Cortical T     positive: cCD3+/CD7+/CD2+/CD5+/CD1a+/CD34+;
                                                                    (T-III)        double positive for CD4/CD8
genitors expressing various patterns of immature T-cell sur-
                                                                    Medullary T    cCD3+/CD7+/CD2+/CD5+/CD1a−/CD34−/
face markers [2, 169, 170]. Precursor T-cell neoplasia with         (T-IV)         membrane CD3+; either positive for CD4 or CD8
predominant mass forming lesions without or with minimal            Early T-ALL    CD7+/CD2+/cCD3+/CD4±, negative for CD1a and
bone marrow involvement are classified as T-lymphoblastic                          CD8, positive for one or more of the myeloid/stem
lymphoma (T-LBL). If these neoplasms are accompanied by                            cell markers CD34, CD117, HLADR, CD13, CD33,
                                                                                   CD11b, CD65
>25% bone marrow blasts, they are generally referred to as
                                                                    Modified according to Béné et al. [171], Arber et al. [2]
T-ALL [169]; see Table 13.10).
may express CD4 but lack CD1a and CD8, and are positive for      as TAL1, TAL2, LYL1, LMO1, LMO2, TLX1, TLX3, ZEB2,
one or more of the myeloid/stem cell markers CD34, CD117,        MYB, and MYC are common in non-ETP T-ALL [19].
HLADR, CD13, CD33, CD11b, or CD65. Myeloperoxidase,              Mutations of LMO genes or TAL1 at 1p32 are sometimes
however, should be negative [2]. The differential diagnosis      associated with MYC translocations [176].
includes especially acute leukemia of ambiguous lineage, espe-      In ETP-ALL mutations in genes that are more commonly
cially T/myeloid mixed phenotype acute leukemia [175].           associated with myeloid neoplasms such as FLT3, NRAS/
                                                                 KRAS, DNMT3A, IDH1/2, RUNX1, and/or ETV6 are found
                                                                 [19, 169].
13.13.3     Genetics
Case History 1: Pro-T-ALL (Fig. 13.47)                                     findings were as follows: expression of cCD3, CD7, CD38,
A 27-year-old woman was admitted with fever and leuko-                     CD45 low, negativity for CD1a, sCD3, CD2, CD4, CD8
cytosis. The CBC was: WBC 53 × 109/L, HB 7.5 g/dL,                         CD13, CD14, CD19, CD34, CD33, CD117, HLA-DR,
platelets 89 × 109/L. The peripheral blood and bone mar-                   TCR alpha/beta, TCR gamma/delta, and TdT. Genetic
row smears as well as the bone marrow core biopsy showed                   analysis revealed a translocation t(14q11.2) involving the
abundant small- sized lymphoblasts with small nucleoli                    α- and δ-T-cell receptor loci. The diagnosis was
and irregular cytoplasmic contours. Immunophenotypic                       pro-T-ALL.
a b
MGG Giemsa
c d
Giemsa cCD3
Fig. 13.47 Pro-T-ALL. (a)Peripheral blood showing numerous small-         blasts (b, c, Giemsa) that express cCD3 (d), CD99 (e), but lack CD1a
sized blasts with slightly indented nuclei, small nucleoli, open chroma-   (f), and are double negative for CD4 (g) and CD8 (h) while some non-
tin, and a small rim of cytoplasm with some protrusions (MGG). (b–h)       neoplastic lymphocytes are positive for CD8. Note nuclear convolution
Bone marrow core biopsy with a diffuse infiltration by small lympho-       of blast cells in (d)
502                      13   Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies
e f
CD99 CD1a
g h
CD4 CD8
Case History 2: Pre-T ALL (Fig. 13.48)                               nearly completely replacing the normal hematopoiesis. By
A 16-year-old male patient was admitted with abdominal pain          multiparametric flow cytometry, the blasts were double-nega-
and circulatory shock. Spontaneous splenic rupture was diag-         tive for CD4 and CD8 but expressed cCD3, CD2, CD5, CD7,
nosed and a splenectomy performed. The CBC was: WBC                  CD38, CD45, and partially CD34. They were negative for
79 × 109/L, HB 5.4 g/dL, platelets 110 × 109/L. The differen-        sCD3, CD13, CD14, CD19, CD33, CD79a. TCR alpha/beta,
tial count showed 64% myeloperoxidase-negative small- to             TCR gamma/delta, HLA-DR, TdT, and myeloperoxidase. The
medium-sized blasts, 2% metamyelocytes, 3% band forms,               spleen was enlarged and showed an infiltration of the red pulp
14% neutrophils, 4% eosinophils, 2% monocytes, and 11%               by cCD3+ blast cells surrounding residual CD79a+ follicles.
lymphocytes. The bone marrow aspirate smear and the bone             Molecular analysis revealed a SIL-TAL1 fusion transcript.
marrow core biopsy revealed a diffuse infiltration by blasts         BCR-ABL1 was negative. The diagnosis was pre-T ALL.
a b
MGG MGG
c d
Giemsa cCD3
Fig. 13.48 Pre-T ALL. (a) Peripheral blood smear with the evidence   biopsy provide evidence of strong cytoplasmic expression of cCD3 (d)
of high leukocytosis (MGG). (b) Bone marrow smears (MGG) and (c)     and CD7 (e). (f–h) Sections cut from the splenectomy specimen (f,
bone marrow core biopsy (Giemsa) showing abundant small- to          H&E) provide evidence of an expansion of the cCD3+ blast cell popu-
medium-sized     blasts  with    prominent    nucleoli.   (d,   e)   lation in the red pulp (g), adjacent to residual CD79a+follicles (h)
Immunohistochemical stains performed on the bone marrow core
504                      13   Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies
e f
CD7 H&E
g h
cCD3 CD79a
Case History 3: Pre-T-ALL with Aberrant Antigen                         little cytoplasm. The immunophenotype was characterized
Expression (Fig. 13.49)                                                 by strong expression of CD2, CD7, cCD3, CD34, TdT, the
A 19-year-old male patient complaint about shortness of                 myeloid-associated antigen CD33, and partially the B-typical
breath. Physical examination revealed a mediastinal mass                antigen CD79a. CD4 and CD8 were negative. Molecular
and pleural effusions. The CBC was: WBC 91 × 109/L, HB                  testing revealed a NOTCH1 activating mutation. The
9.3 g/dL, platelets 2 × 109/L. Peripheral blood, bone marrow            diagnosis was T-ALL of immature thymocyte differentiation
aspirate smear, and trephine bone marrow core biopsy                     corresponding to a pre-T-ALL with aberrant antigen
yielded a diffuse infiltration by small-sized small blasts with          expression.
a b
Giemsa CD7
c d
CD34 CD33
Fig. 13.49 Pre-T-ALL with aberrant antigen expression. (a–h) Sections   CD33 (d). The blast cell population was negative for myeloperoxidase
from the bone marrow core biopsy showing a diffuse infiltration by      that is expressed in some residual myeloid cells (e) but expressed CD99
small-sized blasts (a) Giemsa, strongly expressing CD7 (b), CD34 (c),   (f), TdT (g) and was partially labeled by CD79a antibody (h)
506                      13   Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies
e f
MPO CD99
g h
TdT CD79a
Case History 4: TdT-Negative T-ALL with Partial                             and negative for CD1a, CD4, CD8, TdT, CD117, and myelo-
Expression of CD30 (Fig. 13.50)                                             peroxidase. A bone marrow core biopsy and a soft tissue
A 18-year-old male patient presented with feeling of dizziness,             showed an infiltration by cCD3+, CD99+, CD34+ blasts with
hepatosplenomegaly, and lymphadenopathy. The CBC was:                       a high Ki67+ proliferation fraction. Interestingly, the blast cell
WBC 22 × 109/L, HB 12.8 g/dL, platelets 118 × 109/L. Small-                 population partially expressed CD30 and only faintly expressed
to medium-sized blast cells accounted for 57% of peripheral                 TdT in <5%. Therefore, the neoplasm was considered to be
blood cells. Cerebrospinal fluid puncture revealed a low per-               TdT negative. By FISH, a deletion of 9p21 consistent with a
centage of identical blasts. By multicolor flow immunopheno-                loss of the CDKN2A gene was detected.
typing blasts were positive for cCD3, CD2, CD5, CD7, CD99,                     The diagnosis was TdT-negative pre-T-ALL.
a b
MGG MGG
c d
cCD3 CD99
Fig. 13.50 TdT-negative pre-T-ALL. (a) Blast cells with slightly con-       Ki-67+ proliferation fraction (f). A subpopulation is positive for CD30
voluted nuclei in the cerebrospinal fluid (MGG). (b) Infiltration of adi-   (g) while a faint nuclear staining for TdT (h) is observed in <5% and
pose tissue adjacent to a lymph node by blasts (Giemsa). The neoplastic     therefore considered to be negative
cells express cCD3 (c), CD99 (d), and CD34 (e) and show a high
508                      13   Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies
e f
CD34 Ki67
g h
CD30 TdT
Case History 5: Cortical T-Acute Lymphoblastic                            rim of cytoplasm. The bone marrow core biopsy was hyper-
Leukemia/Lymphoma (Fig. 13.51)                                            cellular and diffusely infiltrated by blasts that expressed
A 39-year-old woman presented with rapidly progressive                    CD3, CD4, CD7, CD8, and CD1a. A phenotypically identi-
shortness of breath. CT scan revealed a large mediastinal that            cal neoplastic population was detected in a needle biopsy
was associated with leukocytosis. The CBC was: WBC                        obtained from the mediastinal mass (Fig. 13.51). However,
79 × 109/L, HB 7.2 g/dL, platelets 42 × 109/L. Peripheral                 the expression of CD1a obtained by immunohistochemistry
blood and bone marrow smears contained about 95%                          was less pronounced than the result obtained by flow from
medium-sized blasts with nuclear convolution and a small                  fresh cells.
a b
H&E CD3
c d
CD4 CD8
Fig. 13.51 Cortical T-ALL/lymphoblastic lymphoma. (a–h) The nee-          CD4 (c) and CD8 (d), partial positivity for CD1a (e), expression of
dle biopsy obtained from the mediastinal mass of the patient showed a     CD99 (f), generally weak expression of CD79a (g) while PAX5 was
diffuse infiltration of medium-sized blast cells with slightly indented   only positive in rare residual B-lymphocytes (h)
nuclei (a, H&E), strong expression of CD3 (b), double reactivity for
510                      13   Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies
e f
CD1a CD99
g h
CD79a PAX5
Case History 6: Early T-Cell Precursor Lymphoblastic                     revealed an expression of CD2, CD7, CD34, CD117, CD38,
Leukemia (Fig. 13.52)                                                    HLA-DR, and a partial expression of cCD3, CD13, and CD33.
An 18-year-old male patient was admitted with fever and                     The blasts were negative for CD1a, CD4, CD5, CD8,
generalized lymphadenopathy. The CBC was: WBC                            CD10, CD14, CD56, lysozyme, and myeloperoxidase. The
31 × 109/L, HB 10.1 g/dL, platelets 73 × 109/L. The LDH                  trephine bone marrow core biopsy was diffusely infiltrated
was 616 U/L. The differential count was as follows: blast                by blasts with an identical immunophenotype. Granulopoiesis
cells 78%, band forms 3%, neutrophils 4%, monocytes 4%,                  was nearly absent while megakaryocytes were only slightly
lymphocytes 11%.                                                         diminished. FISH analysis revealed a TCR α/δ (14q11.2)-
   Peripheral blood and bone marrow smears showed small-                 rearrangement. By next-generation sequencing, a TET2 and
to medium-sized myeloperoxidase-negative blast with incon-               biallelic FLT3 TKD mutations were detected. The diagnosis
spicuous nucleoli and a pale cytoplasm. Flow cytometry                   was early T-cell precursor lymphoblastic leukemia.
a b
MGG MGG
c d
Giemsa Giemsa
Fig. 13.52 Early T-cell precursor lymphoblastic leukemia. (a, b)         (MGG). (c–h) Threphine core bone marrow biopsy shows a dense infil-
Cytologic aspect of peripheral blood (a) and bone marrow smears (b)      tration by blasts with irregularly shaped nuclei (c, d, Giemsa).
showing abundant small- to medium-sized lymphoblasts with incon-         Lymphoblasts strongly express CD7 (e), CD34 (f), CD117 (g), and par-
spicuous nucleoli. No cytoplasmic granulation or Auer rods are present   tially CD33 (h). Note the presence of some residual megakaryocytes
512                      13   Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies
e f
CD7 CD34
g h
CD117 CD33
13.14 P
       recursor Lymphoid Neoplasms:                                Table 13.11 Subtypes of B-lymphoblastic leukemia/lymphoma
      B-Lymphoblastic Leukemia/                                      B-lymphoblastic leukemia/lymphoma, NOS
      Lymphoma                                                       B-lymphoblastic leukemia/lymphoma with recurrent genetic
                                                                      abnormalities
                                                                     B-lymphoblastic leukemia/lymphoma with t(9;22)(q34.1;q11.2);
B-lymphoblastic leukemia/lymphoma B-ALL/LBL is a malig-               BCR-ABL1
nancy of precursor cells of the B-cell lineage generally present-    B-lymphoblastic leukemia/lymphoma with t(v;11q23.3); KMT2A
ing with bone marrow and blood involvement. Primary                   rearranged
extramedullary tumor forming manifestation as B-lymphoblastic        B-lymphoblastic leukemia/lymphoma with t(12;21)(p13.2;q22.1);
lymphoma is rare. B-ALL/LBL is subdivided into multiple enti-         ETV6-RUNX1
                                                                     B-lymphoblastic leukemia/lymphoma with hyperdiploidy
ties according to the genetic background (see Table 13.11) [178].
                                                                     B-lymphoblastic leukemia/lymphoma with hypodiploidy
                                                                     B-lymphoblastic leukemia/lymphoma with t(5;14)(q31.1;q32.3)
                                                                      IL3-IGH
13.14.1     Epidemiology and Clinical Findings                      B-lymphoblastic leukemia/lymphoma with t(1;19)
                                                                      (q23;p13.3);TCF3-PBX1
The estimated annual incidence of ALL is about 1–4.75 cases          Provisional entity: B-lymphoblastic leukemia/lymphoma,
                                                                      BCR-ABL1-like
per 100,000 population [178]. About 75% of cases are diag-           Provisional entity: B-lymphoblastic leukemia/lymphoma with
nosed in children <6 years of age. B-All is much more frequent        iAMP21
than T-ALL and accounts for up to 85% of ALL cases. By con-         From Swerdlow et al. [177]
trast, lymphoblastic lymphoma is more frequently composed
of precursor T-cells than precursor B-cells [178]. The incidence
of B-ALL in infants, children, adolescents, and adults is vari-     Table 13.12 Features of B-ALL of different maturation stages
able according to the different subtypes (see Table 13.11).                            Immunophenotype
    The clinical symptoms are generally related to bone mar-                           Always CD19+ and/or
                                                                    Subtype            CD79a+ and/or CD22+            Cytogenetics
row involvement and replacement of the hematopoietic
                                                                    Early precursor    CD10−                          KMT2A (MLL)
lineages by malignant cells resulting in anemia, granulocyto-       B-ALL (Pro-B)                                     rearrangement
penia, and thrombopenia. Patients may complain about                Common type        CD10+                          All types
bone pain and arthralgia. Lymphadenopathy and hepato-               B-ALL
splenomegaly may develop. B-LBL may be asymptomatic.                Pre-B-ALL          Cytoplasmic μ+
By definition, bone marrow infiltration in B-LBL is <25%.           Transitional       Surface μ+; light chain−
                                                                    pre-B-ALL
                                                                    Mature precursor   Cytoplasmic μ heavy chain+,    t(1;19)
                                                                    B-ALL              cytoplasmic or surface kappa
13.14.2     Morphology and Immunophenotype                                            or lambda; often CD34−
13.14.3      Genetics                                           represented in Hispanics and adult male patients with Down
                                                                 syndrome and associated with inherited genetic variants in
Cytogenetic and molecular findings are essential for the risk    GATA3 [179]. In Ph-like ALL/LBL, multiple genomic altera-
stratification of B-lineage ALL patients (see Table 13.13).      tions may be present that activate kinase and cytokine recep-
Therefore, adequate genetic analysis by karyotyping, FISH,       tor signaling. The genomic landscape varies across the age,
and gene expression profiling is important to determine prog-    clinical features, outcomes, and genetic risk factors [180].
nosis and predictive factors in individual patients [19, 176].
BCR-ABL-like ALL/LBL is characterized by a gene expres-
sion profile similar to that of Ph-positive ALL but lacks the    13.14.4     Caution
BCR-ABL1 translocation [178, 179]. This subtype of
B-precursor neoplasms occurs in all age groups from child-       • Burkitt lymphoma/leukemia is not included into this
hood to old age with prevalence in young adults. It is over-       category.
                                                                 • CD79a is not restricted to B-cells but may also be
                                                                   expressed in specific for a B-lineage derivation but may
Table 13.13 Prognostic groups of B-ALL/LBL according to some       also be expressed in T-ALL/LBL and in some cases of
genetic alterations                                                AML.
Favorable                          Adverse                       • The blastoid variant of mantle cell lymphoma may resem-
High hyperdiploidy (51–65          Hypodiploidy (≤45               ble B-ALL but is TdT negative and generally express
chromosomes)                       chromosomes)                    cyclin D1 or SOX11.
t(12;21)(p13;q22); ETV6-RUNX1      BCR-ABL1                      • Hematogones are B-precursors that may be detected espe-
                                   BCR-ABL1-like
                                                                   cially in bone marrow specimens from infants especially
                                   t(1;19)
                                   iAmp 21
                                                                   after chemotherapy. They show a continuum of expres-
                                   t(v;11q23.3); KMT2A             sion of markers of B-cell differentiation. They are com-
                                   rearranged                      posed of both immature TdT and CD34+cells as well as
Modified from Taylor et al. [19]                                   more mature CD20+ B-cells negative for CD34.
13   Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies                                               515
Case History 1: Pro B-ALL with KMT2A (v;11q23)-                     core biopsy was packed with the same small-sized blasts
Rearrangement (Fig. 13.53)                                          expressing CD19, PAX5, and partially CD34 and TdT but
A 70-year-old woman was admitted with hyperleukocytosis,            negative for CD10 and myeloperoxidase. FISH analysis
severe anemia, and thrombocytopenia. The CBC was: WBC               revealed a KMT2A (v;11q23)-rearrangement. The diagnosis
212 × 109/L, HB 7.5 g/dL, platelets 22 × 109/L. Peripheral          was ALL with KMT2A (v;11q23)-rearrangement according
blood and bone marrow smears showed small blast cells               to the 2016 WHO classification. The immunophenotypic
positive for CD19, CD38, CD79a, HLA-DR, CD15 and par-               subtype according to the maturation stage was pro B-ALL
tially for TdT and CD34. Blasts were negative for CD10,             or early precursor B-ALL according to the
MPO, CD7, CD20, CD33, and CD117. The bone marrow                    EGIL-classification.
a b
MGG MGG
c d
Giemsa Giemsa
Fig. 13.53 Pro B-ALL with KMT2A (v;11q23)-rearrangement. (a, b)     tration of the bone marrow core biopsy by lymphoblasts (Giemsa).
High number of small lymphoblasts in the peripheral blood (a) and   Blasts are partially positive for CD34 (e) and TdT (f) and express CD19
bone marrow (b) smears (May-Grünwald-Giemsa). (c, d) Dense infil-   (g) and PAX5 (h)
516                      13   Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies
e f
CD34 TdT
g h
CD19 PAX5
Case History 2: Common B Lymphoblastic Leukemia in                         expressed cy79a, cyCD34, sCD34, sCD33, HLA-DR,
Down Syndrome (Fig. 13.54)                                                 sCD19, sCD1O, sCD22, and cyTdT and were negative for
A 24-year-old woman with Down syndrome (DS) was                            cyCD3 and myeloperoxidase. The karyotype was 47,XX,+21
admitted to the hospital because of pancytopenia. Myeloid                  indicating a constitutional trisomy 21. No other abnormality
leukemia associated with DS was suspected. The CBC was:                    was present. No translocation t(9;22)/BCR-ABL or MLL
WBC 2.8 × 109/L, HB 6.9 g/dL, platelets 29 × 109/L. The                    (11q23)-rearrangement was detected. The bone marrow
differential count showed 8% small-sized myeloperoxidase-                  core biopsy was diffusely infiltrated by small-sized blast
negative agranular blasts with scanty cytoplasma, 3%                       cells that were negative for myeloperoxidase but strongly
myelocytes, 2% band forms, 3% band forms, 4% mono-                         expressed CD34, CD10, CD19, TdT, and PAX5. In addition,
cytes, 23% neutrophils, 5% eosinophils, and 51% lympho-                    a weak co-expression of CD33 was observed. The diagnosis
cytes. By multiparametric flow cytometry, the CD45+ blasts                 was common B-ALL in DS.
a b
MGG Giemsa
c d
Giemsa MPO
Fig. 13.54 Common acute lymphoblastic leukemia in Down syn-                presence of some eosinophils (Giemsa). (d) Myeloperoxidase is
drome. (a) Blood smear illustrates the presence of rare small-sized lym-   expressed in rare residual granulopoietic precursors while the blast
phoblasts with fine open chromatin, small nucleoli, and a small rim of     population is completely negative. (e–h) The blasts strongly express
cytoplasm (MGG). (b, c) The bone marrow core biopsy is densely infil-      CD10 (e), CD34 (f), CD19 (g), and TdT (h)
trated by blast cells that account for >90% of nucleated cells. Note the
518                      13   Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies
e f
CD10 CD34
g h
CD19 TdT
Case History 3: B-Lymphoblastic Leukemia with t(9;22)                       cCD3, CD11c, CD15, CD33, cIgM, and myeloperoxidase.
(q34.1;q11.2);BCR-ABL1 (Fig. 13.55)                                         Bone marrow aspirate smears and the core biopsy showed a
A 37-year-old woman developed bone pain and fever. The CBC                  dense infiltration by lymphoblasts. Karyotyping and FISH anal-
was: WBC 56 × 109/L, HB 6.3 g/dL, platelets 48 × 109/L. The                 ysis revealed a t(9;22)(q34.1;q11.2); BCR-ABL1. By multiplex
peripheral blood contained 85% medium-sized blasts that were               PCR, a BCR-ABL minor fusion transcript was detected. The
positive for CD19, cCD22, cCD79a, CD10, HLA DR, and TdT                     diagnosis was B-lymphoblastic leukemia with t(9;22)
and partially positive for CD20 and CD13 but negative for                   (q34.1;q11.2); BCR-ABL1.
a b
MGG Giemsa
c d
Giemsa CD19
Fig. 13.55 B-lymphoblastic leukemia with t(9;22)(q34.1;q11.2);              and a pale basophilic cytoplasm. Note the presence of scattered pro-
BCR-ABL1. (a) A peripheral blood smear showing abundant medium-            erythroblasts in b (Giemsa). (d) The strongly CD19+ lymphoblasts sur-
sized lymphoblasts with slightly irregular nuclei and evenly dispersed      round a residual CD19-negative megakaryocyte. (e–h) The neoplastic
chromatin (May-Grünwald-Giemsa). (b, c) The trephine bone marrow            cells also express CD10 (e), PAX5 (f), CD34 (g), and TdT (h)
core biopsy is densely infiltrated by blast cells with prominent nucleoli
520                      13   Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies
e f
CD10 PAX5
g h
CD34 TdT
Case History 4: Mature B-Lymphoblastic Leukemia                             CD19, CD20, CD79a, surface Kappa-light chain, and
with TP53 Mutation (Fig. 13.56)                                             IgM. They were partially positive for TdT and CD34 but
A 54-year-old woman developed hepatosplenomegaly, rap-                      were negative for CD10, myeloperoxidase, CD33, and
idly progressing lymphadenopathy, and fever. The CBC was:                   CD117. Genetic analysis showed a TP53 mutation indicating
WBC 48 × 109/L, HB 10.2 g/dL, platelets 19 × 109/L. The                     a high-risk group. In contrast to Burkitt lymphoma/leuke-
peripheral blood and bone marrow smears showed about                        mia, no MYC translocation was detected.
90% small- to medium-sized blasts that were positive for                       The diagnosis was mature B-ALL with TP53 mutation.
a b
Giemsa Giemsa
c d
CD79a CD10
Fig. 13.56 Mature B-lymphoblastic leukemia with TP53 mutation.              for CD10 (d). Blasts expressed CD20 (e), IgM (f), and were partially
(a–h) Sections cut from bone marrow core biopsy showing a dense             positive for p53 (g). The Ki67+ proliferation fraction was about 98%
infiltration by blasts (a, b, Giemsa) positive for CD79a (c) but negative   (h)
522                      13   Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies
e f
CD20 IgM
g h
p53 Ki67
Case History 5: Pre-B-ALL with Subtle Bone Marrow             aspirate and core biopsy were performed and contained about
Infiltration at Relapse (Fig. 13.57)                          20% blasts that were positive for CD34, CD19, CD22, TdT,
A 44-year-old woman was diagnosed with pre-B- surface μ chain, and partially for CD10 but negative for CD20,
ALL. Treatment by standard chemotherapy achieved complete CD3, CD33, CD117, and myeloperoxidase. In addition, an
remission. When she presented a year later for hematological aberrant expression of CD7 was present.
control, the CBC was: WBC 4.3 × 109/L, HB 12.6 g/dL, plate-      Hematogones were excluded because of the homogeneous
lets 204 × 109/L. However, cytologic evaluation of peripheral immunophenotype and the aberrant expression of CD7 by
blood smears revealed rare myeloperoxidase-negative blasts B-lineage blasts. The diagnosis was relapse of a pre-B-ALL
accounting for about 4% of nucleated cells. A bone marrow with subtle bone marrow and peripheral blood involvement.
a b
MGG MGG
c d
Giemsa Giemsa
Fig. 13.57 Pre-B-ALL with subtle bone marrow infiltration at relapse.       showing scattered blasts between hematopoietic precursors (Giemsa).
Rare lymphoblasts in a peripheral blood (a) and bone marrow aspirate (b)    Immunohistochemical staining for CD19 (e), CD10 (f), CD34 (g), and TdT
smears (May-Grünwald-Giemsa). Note discrete signs of dyshematopoiesis       (h) highlights interstitial bone marrow infiltration by B-lymphoblastic leu-
after chemotherapy. (c, d) Slightly hypercellular bone marrow core biopsy   kemia without significant replacement of hematopoiesis
524                      13   Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies
e f
CD19 CD10
g h
CD34 TdT
Case History 6: Pro-B-ALL with Partial Spontaneous                     aspirate smear was hypocellular and showed fat cells and
Bone Marrow Necrosis in a Patient with Down                            cellular debris. A bone marrow core biopsy was performed
Syndrome (Fig. 13.58)                                                  and revealed extensive bone marrow necrosis. Adjacent
A 43-year-old man with Down syndrome was admitted                      areas were infiltrated by small-sized blasts strongly express-
with pneumonia, bone pain, and tricytopenia. The CBC                   ing CD79a and TdT but negative for CD10. The necrotic
was: WBC 2.1 × 109/L, HB 6.1 g/dL, platelets                           areas were weakly immunolabeled by CD79a antibody. The
12 × 109/L. The differential count showed 30% neutrophils,             diagnosis was pro-B-ALL (early B-ALL) with partial spon-
5% band forms, 2% metamyelocytes, 1% monocytes, 59%                    taneous bone marrow necrosis in a patient with Down
lymphocytes, and 3% small-sized blasts. Bone marrow                    syndrome.
a b
MGG MGG
c d
Giemsa Giemsa
Fig. 13.58 Pro-B-ALL with partial bone marrow necrosis in a patient    of necrosis (c, d, Giemsa) and areas densely infiltrated by blasts (c, e,
with Down syndrome. (a) A rare peripheral blood lymphoblast with a     f). (g) CD79a is strongly expressed by blasts that weakly stain the
high nuclear:cytoplasmic ratio, condensed chromatin, and inconspicu-   necrotic areas. (h) Strong nuclear immunolabeling of non-necrotic blast
ous nucleoli (MGG). (b) Bone marrow aspirate showing fat cells and     cells by TdT antibody
nuclear debris (MGG). (c–h) Bone marrow core biopsy showing areas
526                      13   Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies
e f
Giemsa Giemsa
g h
CD79a TdT
Acknowledgments The author thanks Dr. Dietmar Pfeifer, Dr. Milena                  H, Thiele J, editors. WHO classification. Revised 4 ed. Lyon: IARC
Pantic and Dr. Justyna Rawluk, Department of Medicine I, Medical                   Press; 2017. p. 150–2.
Center – University of Freiburg, Germany for helpfull analysis and dis-      13.   Vardiman JW, Arber DA, Brunning RD, Larson RA, Matutes E,
cussion of molecular genetic, cytogenetic, cytologic and immunophe-                Baumann I, Kvasnicka HM. Therapy-related myeloid neoplasms.
notypic data.                                                                      In: Swerdlow SH, Campo E, Haris NL, Jaffe ES, Pileri SA, Stein
                                                                                   H, Thiele J, editors. WHO classification. Revised 4 ed. Lyon: IARC
                                                                                   Press; 2017. p. 153–5.
                                                                             14.   Arber DA, Brunning RD, Orazi A, Porwit A, Peterson LC, Thiele J,
                                                                                   Le Beau MM, Hassedjian RP. Acute myeloid leukemia, NOS. In:
References                                                                         Swerdlow SH, Campo E, Haris NL, Jaffe ES, Pileri SA, Stein H,
                                                                                   Thiele J. WHO classification. Revised 4 IARC Press, Lyon 2017.
 1. Kampen KR. The discovery and early understanding of leuke-                     pp 156–66.
    mia. Leuk Res. 2012;36(1):6–13. https://doi.org/10.1016/j.leu-           15.   Pileri SA, Orazi A, Falini P. Myeloid sarcoma. In Swerdlow SH,
    kres.2011.09.028. Epub 2011 Oct 26.                                            Campo E, Haris NL, Jaffe ES, Pileri SA, Stein H, Thiele J. WHO
 2. Arber DA, Orazi A, Hasserjian R, Thiele J, Borowitz MJ, Le Beau                classification. Revised 4 pp 167–168, IARC Press, Lyon 2017.
    MM, Bloomfield CD, Cazzola M, Vardiman JW. The 2016 revi-                16.   Arber DA, Baumann I, Niemeyer CM, Brunning RD, Porwit
    sion to the World Health Organization classification of myeloid                A. Myeloid proliferations associated with Down syndrome. In:
    neoplasms and acute leukemia. Blood. 2016;127(20):2391–405.                    Swerdlow SH, Campo E, Haris NL, Jaffe ES, Pileri SA, Stein H,
    https://doi.org/10.1182/blood-2016-03-643544. Epub 2016 Apr 11.                Thiele J, editors. WHO classification. Revised 4 ed. Lyon: IARC
    Review.                                                                        Press; 2017. p. 169–71.
 3. Kurzer JH, Weinberg OK. Acute Leukemias of Ambiguous                     17.   Papaemmanuil E, Gerstung M, Bullinger L, Gaidzik VI, Paschka
    Lineage: Clarification on Lineage Specificity. Surg Pathol Clin.               P, Roberts ND, Potter NE, Heuser M, Thol F, Bolli N, Gundem
    2019;12(3):687–97.         https://doi.org/10.1016/j.path.2019.03.008.         G, Van Loo P, Martincorena I, Ganly P, Mudie L, McLaren S,
    Review.                                                                        O’Meara S, Raine K, Jones DR, Teague JW, Butler AP, Greaves
 4. Crispino JD, Horwitz MS. GATA factor mutations in hematologic                  MF, Ganser A, Döhner K, Schlenk RF, Döhner H, Campbell
    disease. Blood. 2017;129(15):2103–10. https://doi.org/10.1182/                 PJ. Genomic classification and prognosis in acute myeloid
    blood-2016-09-687889. Epub 2017 Feb 8.                                         leukemia. N Engl J Med. 2016;374(23):2209–21. https://doi.
 5. Niemeyer CM, Mecucci C. Practical considerations for diagno-                   org/10.1056/NEJMoa1516192.
    sis and management of patients and carriers. Semin Hematol.              18.   Grimwade D, Ivey A, Huntly BJ. Molecular landscape of acute
    2017;54(2):69–74.                https://doi.org/10.1053/j.seminhema-          myeloid leukemia in younger adults and its clinical relevance.
    tol.2017.04.002. Epub 2017 Apr 6.                                              Blood. 2016;127(1):29–41.
 6. Döhner H, Estey E, Grimwade D, Amadori S, Appelbaum FR,                  19.   Taylor J, Xiao W, Abdel-Wahab O. Diagnosis and classification
    Büchner T, Dombret H, Ebert BL, Fenaux P, Larson RA, Levine                    of hematologic malignancies on the basis of genetics. Blood.
    RL, Lo-Coco F, Naoe T, Niederwieser D, Ossenkoppele GJ,                        2017;130(4):410–23.             https://doi.org/10.1182/blood-2017-
    Sanz M, Sierra J, Tallman MS, Tien HF, Wei AH, Löwenberg B,                    02-734541. Epub 2017 Jun 9.
    Bloomfield CD. Diagnosis and management of AML in adults:                20.   Döhner H, Estey EH, Amadori S, Appelbaum FR, Büchner T,
    2017 ELN recommendations from an international expert panel.                   Burnett AK, Dombret H, Fenaux P, Grimwade D, Larson RA,
    Blood. 2017;129(4):424–47. https://doi.org/10.1182/blood-2016-                 Lo-Coco F, Naoe T, Niederwieser D, Ossenkoppele GJ, Sanz MA,
    08-733196. Epub 2016 Nov 28. Review.                                           Sierra J, Tallman MS, Löwenberg B, Bloomfield CD, European
 7. Weinberg OK, Sohani AR, Bhargava P, Nardi V. Diagnostic work-                  LeukemiaNet. Diagnosis and management of acute myeloid leuke-
    up of acute myeloid leukemia. Am J Hematol. 2017;92(3):317–21.                mia in adults: recommendations from an international expert panel,
     https://doi.org/10.1002/ajh.24648. Epub 2017 Feb 3. Review.                   on behalf of the European LeukemiaNet. Blood. 2010;115(3):453–
 8. Arber DA, Borowitz MJ, Cessna M, Etzell J, Foucar K, Hasserjian                74. https://doi.org/10.1182/blood-2009-07-235358. Epub 2009 Oct
     RP, Rizzo JD, Theil K, Wang SA, Smith AT, Rumble RB,                          30.
     Thomas NE, Vardiman JW. Initial diagnostic workup of acute              21.   Gerstung M, Papaemmanuil E, Martincorena I, Bullinger L,
     leukemia: guideline from the College of American Pathologists                 Gaidzik VI, Paschka P, Heuser M, Thol F, Bolli N, Ganly P, Ganser
     and the American Society of Hematology. Arch Pathol Lab Med.                  A, McDermott U, Döhner K, Schlenk RF, Döhner H, Campbell
     2017; https://doi.org/10.5858/arpa.2016-0504-CP. [Epub ahead                  PJ. Precision oncology for acute myeloid leukemia using a
     of print].                                                                    knowledge bank approach. Nat Genet. 2017;49(3):332–40. https://
 9. Bennett JM, Catovsky D, Daniel MT, et al. Proposed revised cri-                doi.org/10.1038/ng.3756. Epub 2017 Jan 16.
     teria for the classification of acute myeloid leukemia. A report of     22.   Shysh AC, Nguyen LT, Guo M, Vaska M, Naugler C, Rashid-
     the French-American-British Cooperative Group. Ann Intern Med.                Kolvear F. The incidence of acute myeloid leukemia in Calgary,
     1985;103:626–9.                                                               Alberta, Canada: a retrospective cohort study. BMC Public Health.
10. Vardiman JW, Thiele J, Arber DA, Brunning RD, Borowitz                         2017;18(1):94. https://doi.org/10.1186/s12889-017-4644-6.
     MJ, Porwit A, Harris NL, Le Beau MM, Hellström-Lindberg                 23.   Acute myeloid leukaemia (AML) incidence statistics. London:
     E, Tefferi A, Bloomfield CD. The 2008 revision of the World                   Cancer Research UK; 2016. http://www.cancerresearchuk.org/
     Health Organization (WHO) classification of myeloid neo-                      health-professional/cancer-statistics/statistics-by-cancer-type/
     plasms and acute leukemia: rationale and important changes.                   leukaemia-aml/incidence.
     Blood. 2009;114(5):937–51. https://doi.org/10.1182/blood-2009-          24.   Deschler B, Lübbert M. Acute myeloid leukemia: epidemiology
     03-209262. Epub 2009 Apr 8. Review.                                          and etiology. Cancer. 2006;107(9):2099–107. Review.
11. Arber DA, Brunning RD, Le Beau MM, Falini B, Vardiman JW,                25.   Zwaan CM, Kolb EA, Reinhardt D, Abrahamsson J, Adachi S,
     Porwit A, Thiele J, Foucar K, Döhner H, Bloomfield CD. AML                    Aplenc R, De Bont ES, De Moerloose B, Dworzak M, Gibson BE,
     with recurrent genetic abnormalities. In: Swerdlow SH, Campo E,               Hasle H, Leverger G, Locatelli F, Ragu C, Ribeiro RC, Rizzari
     Haris NL, Jaffe ES, Pileri SA, Stein H, Thiele J, editors. WHO clas-          C, Rubnitz JE, Smith OP, Sung L, Tomizawa D, van den Heuvel-
     sification. Revised 4 ed. Lyon: IARC Press; 2017. p. 130–49.                  Eibrink MM, Creutzig U, Kaspers GJ. Collaborative efforts driv-
12. Arber DA, Brunning RD, Orazi A, Bain BJ, Porwit A, Le Beau                     ing progress in pediatric acute myeloid leukemia. J Clin Oncol.
     MM, Greenberg PL. AML with myelodysplasia-related changes.                    2015;33(27):2949–62. https://doi.org/10.1200/JCO.2015.62.8289.
     In: Swerdlow SH, Campo E, Haris NL, Jaffe ES, Pileri SA, Stein                Epub 2015 Aug 24. Review.
528                                            13   Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies
26. Taga T, Tomizawa D, Takahashi H, Adachi S. Acute myeloid leu-           39. Dillon R, Grimwade D. Just 1 test to diagnose AML?!! Blood.
    kemia in children: Current status and future directions. Pediatr Int.       2016;128(1):8–10. https://doi.org/10.1182/blood-2016-05-715060.
    2016;58(2):71–80. Review. https://doi.org/10.1111/ped.12865.            40. Herold T, Rothenberg-Thurley M, Grunwald VV, Janke H, Goerlich
27. O’Donnell MR, Tallman MS, Abboud CN, Altman JK, Appelbaum                   D, Sauerland MC, Konstandin NP, Dufour A, Schneider S, Neusser
    FR, Arber DA, Bhatt V, Bixby D, Blum W, Coutre SE, De Lima M,               M, Ksienzyk B, Greif PA, Subklewe M, Faldum A, Bohlander
    Fathi AT, Fiorella M, Foran JM, Gore SD, Hall AC, Kropf P, Lancet           SK, Braess J, Wörmann B, Krug U, Berdel WE, Hiddemann W,
    J, Maness LJ, Marcucci G, Martin MG, Moore JO, Olin R, Peker D,             Spiekermann K, Metzeler KH. Validation and refinement of the
    Pollyea DA, Pratz K, Ravandi F, Shami PJ, Stone RM, Strickland              revised 2017 European LeukemiaNet genetic risk stratification
    SA, Wang ES, Wieduwilt M, Gregory K, Ogba N. Acute Myeloid                  of acute myeloid leukemia. Leukemia. 2020 Mar 30. https://doi.
    Leukemia, Version 3.2017, NCCN Clinical Practice Guidelines in              org/10.1038/s41375-020-0806-0. Online ahead of print.
    Oncology. J Natl Compr Canc Netw. 2017;15(7):926–57. https://           41. Döhner H, Estey E, Grimwade D, et al. Diagnosis and management
    doi.org/10.6004/jnccn.2017.0116.                                            of AML in adults: 2017 ELN recommendations from an interna-
28. Haferlach T, Winkemann M, Löffler H, Schoch R, Gassmann                     tional expert panel. Blood. 2016;8:733196.
    W, Fonatsch C, Schoch C, Poetsch M, Weber-Matthiesen K,                 42. Kayser S, Schlenk RF, Platzbecker U. Management of patients with
    Schlegelberger B. The abnormal eosinophils are part of the leuke-           acute promyelocytic leukemia. Leukemia. 2018;32(6):1277–94.
    mic cell population in acute myelomonocytic leukemia with abnor-            Epub 2018 Apr 24.
    mal eosinophils (AML M4Eo) and carry the pericentric inversion          43. Baba SM, Pandith AA, Shah ZA, Baba RA. Pathogenetic impli-
    16: a combination of May-Grünwald-Giemsa staining and fluores-              cation of fusion genes in acute promyelocytic leukemia and their
    cence in situ hybridization. Blood. 1996;87(6):2459–63.                     diagnostic utility. Clin Genet. 2019;95(1):41–52. https://doi.
29. Haferlach T, Kohlmann A, Klein HU, Ruckert C, Dugas M,                      org/10.1111/cge.13372. Epub 2018 Jun 8. Review.
    Williams PM, Kern W, Schnittger S, Bacher U, Löffler H, Haferlach       44. Lo-Coco F, Cicconi L, Breccia M. Current standard treat-
    C. AML with translocation t(8;16)(p11;p13) demonstrates unique              ment of adult acute promyelocytic leukaemia. Br J Haematol.
    cytomorphological, cytogenetic, molecular and prognostic fea-               2016;172(6):841–54.
    tures. Leukemia. 2009;23(5):934–43. https://doi.org/10.1038/            45. Cicconi L, Fenaux P, Kantarjian H, Tallman M, Sanz MA, Lo-Coco
    leu.2008.388. Epub 2009 Feb 5.                                              F. Molecular remission as a therapeutic objective in acute pro-
30. Chen W, Konoplev S, Medeiros LJ, Koeppen H, Leventaki V,                    myelocytic leukemia. Leukemia. 2018; https://doi.org/10.1038/
    Vadhan-Raj S, Jones D, Kantarjian HM, Falini B, Bueso-Ramos                 s41375-018-0219-5. Epub ahead of print] Review.
    CE. 18. Cuplike nuclei (prominent nuclear invaginations) in acute       46. Balduini CL. 100-Year-Old Haematologica Images: Acute
    myeloid leukemia are highly associated with FLT3 internal tan-              Promyelocytic Leukemia. Haematologica. 2020 Jan 31;105(2):245.
    dem duplication and NPM1 mutation. Cancer. 2009;115(23):                    https://doi.org/10.3324/haematol.2020.247056. Print 2020.
    5481–9.                                                                 47. Wang HY, Rashidi HH. The new clinicopathologic and molecu-
31. Dimitrienko S, Vercauteren S. Auer rods in mature granulocytes of           lar findings in myeloid neoplasms with inv(3)(q21q26)/t(3;3)
    a patient with mixed lineage leukemia. Blood. 2012;119(19):4348.            (q21;q26.2). Arch Pathol Lab Med. 2016;140(12):1404–10. Epub
32. Béné MC, Nebe T, Bettelheim P, Buldini B, Bumbea H, Kern                    2016 Sep 15.
    W, Lacombe F, Lemez P, Marinov I, Matutes E, Maynadié                   48. Sweet DL, Golomb HM, Rowley JD, Vardiman JM. Acute
    M, Oelschlagel U, Orfao A, Schabath R, Solenthaler M,                       myelogenous leukemia and thrombocythemia associated with
    Tschurtschenthaler G, Vladareanu AM, Zini G, Faure GC, Porwit               an abnormality of chromosome No. 3. Cancer Genet Cytogenet.
    A. Immunophenotyping of acute leukemia and lymphoprolifera-                 1979;1(1):33–7.
    tive disorders: a consensus proposal of the European LeukemiaNet        49. Hinai AA, Valk PJ. Review: aberrant EVI1 expression in acute
    Work Package 10. Leukemia. 2011;25(4):567–74. https://doi.                  myeloid leukaemia. Br J Haematol. 2016;172(6):870–8. https://doi.
    org/10.1038/leu.2010.312. Epub 2011 Jan 21.                                 org/10.1111/bjh.13898. Epub 2016 Jan 5. Review.
33. Buldini B, Rizzati F, Masetti R, Fagioli F, Menna G, Micalizzi C,       50. Baldazzi C, Luatti S, Zuffa E, Papayannidis C, Ottaviani E,
    Putti MC, Rizzari C, Santoro N, Zecca M, Disarò S, Rondelli R,              Marzocchi G, Ameli G, Bardi MA, Bonaldi L, Paolini R, Gurrieri C,
    Merli P, Pigazzi M, Pession A, Locatelli F, Basso G. Prognostic             Rigolin GM, Cuneo A, Martinelli G, Cavo M, Testoni N. Complex
    significance of flow-cytometry evaluation of minimal residual               chromosomal rearrangements leading to MECOM overexpression
    disease in children with acute myeloid leukaemia treated accord-            are recurrent in myeloid malignancies with various 3q abnormali-
    ing to the AIEOP-AML 2002/01 study protocol. Br J Haematol.                 ties. Genes Chromosomes Cancer. 2016;55(4):375–88. https://doi.
    2017;177(1):116–26. https://doi.org/10.1111/bjh.14523. Epub                 org/10.1002/gcc.22341. Epub 2016 Jan 27.
    2017 Feb 27.                                                            51. Gröschel S, Sanders MA, Hoogenboezem R, et al. Mutational
34. Rowley JD. Identification of a translocation with quinacrine fluo-          spectrum of myeloid malignancies with inv(3)/t(3;3) reveals a
    rescence in a patient with acute leukemia. Ann Genet. 1973;16:              predominant involvement of RAS/RTK signaling pathways. Blood.
    109–12.                                                                     2015;125(1):133–9.
35. Rowley JD. Chromosomal translocations: revisited yet                    52. Lugthart S, Gröschel S, Beverloo HB, Kayser S, Valk PJ, van
    again. Blood. 2008;112(6):2183–9. https://doi.org/10.1182/                  Zelderen-Bhola SL, Jan Ossenkoppele G, Vellenga E, van den
    blood-2008-04-097931.                                                       Berg-de Ruiter E, Schanz U, Verhoef G, Vandenberghe P, Ferrant A,
36. Grimwade D, Mrózek K. Diagnostic and prognostic value of                    Köhne CH, Pfreundschuh M, Horst HA, Koller E, von Lilienfeld-
    cytogenetics in acute myeloid leukemia. Hematol Oncol Clin                  Toal M, Bentz M, Ganser A, Schlegelberger B, Jotterand M,
    North Am. 2011;25(6):1135–61, vii. https://doi.org/10.1016/j.               Krauter J, Pabst T, Theobald M, Schlenk RF, Delwel R, Döhner
    hoc.2011.09.018.                                                            K, Löwenberg B, Döhner H. Clinical, molecular, and prognostic
37. Döhner H. Implication of the molecular characterization of acute            significance of WHO type inv(3)(q21q26.2)/t(3;3)(q21;q26.2)
    myeloid leukemia. Hematology Am Soc Hematol Educ Program.                   and various other 3q abnormalities in acute myeloid leukemia.
    2007:412–9.                                                                 J Clin Oncol. 2010;28(24):3890–8. https://doi.org/10.1200/
38. Bullinger L, Döhner K, Döhner H. Genomics of acute myeloid leu-             JCO.2010.29.2771. Epub 2010 Jul 26.
    kemia diagnosis and pathways. J Clin Oncol. 2017;35(9):934–46.          53. Lin S, Mulloy JC, Goyama S. RUNX1-ETO Leukemia.
    https://doi.org/10.1200/JCO.2016.71.2208. Epub 2017 Feb 13.                 Adv      Exp     Med     Biol.    2017;962:151–73.    https://doi.
    Review.                                                                     org/10.1007/978-981-10-3233-2_11.
13   Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies                                                     529
54. Schnittger S, Tobias M, Kohl TM, Torsten Haferlach T, Wolfgang           NUP214-rearranged pediatric myeloid leukemia: an international
     Kern W, Wolfgang Hiddemann W, Karsten Spiekermann K, Claudia            study of 62 patients. Haematologica. 2014;99(5):865–72. https://
     Schoch C. KIT-D816 mutations in AML1-ETO-positive AML are               doi.org/10.3324/haematol.2013.098517. Epub 2014 Jan 17.
     associated with impaired event-free and overall survival. Blood.    66. Foucar K, Anastasi J. Acute myeloid leukemia with recurrent cyto-
     2006;107:1791–9.                                                        genetic abnormalities. Am J Clin Pathol. 2015;144(1):6–18.
55. Park SH, Chi HS, Cho YU, Jang S, Park CJ. Effects of c-KIT muta-     67. Tarlock K, Alonzo TA, Moraleda PP, Gerbing RB, Raimondi SC,
     tions on expression of the RUNX1/RUNX1T1 fusion transcript              Hirsch BA, Ravindranath Y, Lange B, Woods WG, Gamis AS,
     in t(8;21)-positive acute myeloid leukemia patients. Leuk Res.          Meshinchi S. Acute myeloid leukaemia (AML) with t(6;9)(p23;q34)
     2013;37(7):784–9.                                                       is associated with poor outcome in childhood AML regardless of
56. Micol JB, Duployez N, Boissel N, Petit A, Geffroy S, Nibourel            FLT3-ITD status: a report from the Children’s Oncology Group. Br
     O, Lacombe C, Lapillonne H, Etancelin P, Figeac M, Renneville           J Haematol. 2014;166(2):254–9. https://doi.org/10.1111/bjh.12852.
     A, Castaigne S, Leverger G, Ifrah N, Dombret H, Preudhomme C,           Epub 2014 Mar 25.
     Abdel-Wahab O, Jourdan E. Frequent ASXL2 mutations in acute         68. Heuser M. Therapy-related myeloid neoplasms: does knowing the
     myeloid leukemia patients with t(8;21)/RUNX1-RUNX1T1 chro-              origin help to guide treatment? Hematology Am Soc Hematol Educ
     mosomal translocations. Blood. 2014;124(9):1445–9. https://doi.         Program. 2016;2016(1):24–32.
     org/10.1182/blood-2014-04-571018. Epub 2014 Jun 27.                 69. Balgobind BV, Zwaan CM, Pieters R, Van den Heuvel-Eibrink
57. Jahn N, Agrawal M, Bullinger L, Weber D, Corbacioglu A,                  MM. The heterogeneity of pediatric MLL-rearranged acute myeloid
     Gaidzik VI, Schmalbrock L, Thol F, Heuser M, Krauter J, Göhring         leukemia. Leukemia. 2011;25(8):1239–48.
     G, Kündgen A, Fiedler W, Wattad M, Held G, Köhne CH, Horst          70. Bolouri H, Farrar JE, Triche T Jr, Ries RE, Lim EL, Alonzo TA,
     HA, Lübbert M, Ganser A, Schlenk RF, Döhner H, Döhner K,                Ma Y, Moore R, Mungall AJ, Marra MA, Zhang J, Ma X, Liu Y,
     Paschka P. Incidence and prognostic impact of ASXL2 mutations           Liu Y, Auvil JMG, Davidsen TM, Gesuwan P, Hermida LC, Salhia
     in adult acute myeloid leukemia patients with t(8;21)(q22;q22):         B, Capone S, Ramsingh G, Zwaan CM, Noort S, Piccolo SR, Kolb
     a study of the German-Austrian AML Study Group. Leukemia.               EA, Gamis AS, Smith MA, Gerhard DS, Meshinchi S. The molecu-
     2017;31(4):1012–5. https://doi.org/10.1038/leu.2017.18. Epub            lar landscape of pediatric acute myeloid leukemia reveals recurrent
     2017 Jan 16.                                                            structural alterations and age-specific mutational interactions. Nat
58. Johnson RC, Ma L, Cherry AM, Arber DA, George TI. B-cell tran-           Med. 2018;24(1):103–12. https://doi.org/10.1038/nm.4439.
     scription factor expression and immunoglobulin gene rearrange-      71. Meyer C, Burmeister T, Gröger D, Tsaur D, Fechina L, et al.
     ment frequency in acute myeloid leukemia with t(8;21)(q22;q22).         The MLL recombinome of acute leukemias in 2017. Leukemia.
     Am J Clin Pathol. 2013;140:355–62.                                      2018;32:273–84. https://doi.org/10.1038/leu.2017.213.
59. Grimwade D, Walker H, Oliver F, et al. The importance of diag-       72. Gröschel S, Schlenk RF, Engelmann J, Rockova V, Teleanu V, Kühn
     nostic cytogenetics on outcome in AML: analysis of 1,612 patients       MW, et al. Deregulated expression of EVI1 defines a poor prognos-
     entered into the MRC AML 10 trial. Blood. 1998;92:2322–33.              tic subset of MLL-rearranged acute myeloid leukemias: a study of
60. Delaunay J, Vey N, Leblanc T, Fenaux P, Rigal-Huguet F, Witz             the German-Austrian Acute Myeloid Leukemia Study Group and
     F, Lamy T, Auvrignon A, Blaise D, Pigneux A, Mugneret F,                the Dutch-Belgian-Swiss HOVON/SAKK Cooperative Group. J
     Bastard C, Dastugue N, Van den Akker J, Fière D, Reiffers J,            Clin Oncol. 2013;31(1):95–103.
     Castaigne S, Leverger G, Harousseau JL, Dombret H. Prognosis        73. Kühn MW, Bullinger L, Gröschel S, Krönke J, Edelmann J, Rücker
     of inv(16)/t(16;16) acute myeloid leukemia (AML): a sur-                FG, Eiwen K, Paschka P, Gaidzik VI, Holzmann K, Schlenk RF,
     vey of 110 cases from the French AML Intergroup. Blood.                 Döhner H, Döhner K. Genome-wide genotyping of acute myeloid
     2003;102(2):462–9.                                                      leukemia with translocation t(9;11)(p22;q23) reveals novel recur-
61. Salem A, Loghavi S, Tang G, Huh YO, Jabbour EJ, Kantarjian H,            rent genomic alterations. Haematologica. 2014;99(8):e133–5.
     Wang W, Hu S, Luthra R, Medeiros LJ, Khoury JD. Myeloid neo-        74. Gruber TA, Downing JR. The biology of pediatric acute mega-
     plasms with concurrent BCR-ABL1 and CBFB rearrangements:                karyoblastic leukemia. Blood. 2015;126(8):943–9. https://doi.
     A series of 10 cases of a clinically aggressive neoplasm. Am J          org/10.1182/blood-2015-05-567859.
     Hematol. 2017;92(6):520–8.                                          75. Teyssier AC, Lapillonne H, Pasquet M, Ballerini P, Baruchel
62. Rogers HJ, Hsi ED, Tang G, Wang SA, Bueso-Ramos CE, Lubin D,             A, Ducassou S, Fenneteau O, Petit A, Cuccuini W, Ragu C,
     Morrissette JJ, Bagg A, Cherukuri DP, George TI, Peterson L, Liu        Preudhomme C, Mercher T, Sirvent N, Leverger G. Acute mega-
     YC, Mathew S, Orazi A, Hasserjian RP. Most myeloid neoplasms            karyoblastic leukemia (excluding Down syndrome) remains an
     with deletion of chromosome 16q are distinct from acute myeloid         acute myeloid subgroup with inferior outcome in the French
     leukemia with Inv(16)(p13.1q22): A Bone Marrow Pathology                ELAM02 trial. Pediatr Hematol Oncol. 2017;34(8):425–7. https://
     Group Multicenter Study. Am J Clin Pathol. 2017;147(4):411–9.           doi.org/10.1080/08880018.2017.1414905.
63. Visconte V, Shetty S, Przychodzen B, Hirsch C, Bodo J,               76. Klairmont MM, Hoskoppal D, Yadak N, Choi JK. The comparative
    Maciejewski JP, Hsi ED, Rogers HJ. Clinicopathologic and                 sensitivity of immunohistochemical markers of megakaryocytic dif-
    molecular characterization of myeloid neoplasms with isolated            ferentiation in acute megakaryoblastic leukemia. Am J Clin Pathol.
    (6;9)(p23;q34). Int J Lab Hematol. 2017;39(4):409–17. https://           2018; https://doi.org/10.1093/ajcp/aqy074. [Epub ahead of print].
    doi.org/10.1111/ijlh.12641.                                          77. Carroll A, Civin C, Schneider N, et al. The t(1;22) (p13;q13)
64. Ommen HB, Touzart A, MacIntyre E, Kern W, Haferlach T,                   is nonrandom and restricted to infants with acute megakaryo-
    Haferlach C, Tobal K, Hokland P, Schnittger S. The kinetics of           blastic leukemia: a Pediatric Oncology Group Study. Blood.
    relapse in DEK-NUP214-positive acute myeloid leukemia patients.          1991;78(3):748–52.
    Eur J Haematol. 2015;95(5):436–41. https://doi.org/10.1111/          78. Alford KA, Reinhardt K, Garnett C, et al.; International Myeloid
    ejh.12511. Epub 2015 Mar 13.                                             Leukemia-Down Syndrome Study Group Analysis of GATA1 muta-
65. Sandahl JD, Coenen EA, Forestier E, Harbott J, Johansson B,              tions in Down syndrome transient myeloproliferative disorder and
    Kerndrup G, Adachi S, Auvrignon A, Beverloo HB, Cayuela JM,              myeloid leukemia. Blood. 2011;118(8):2222-2238.
    Chilton L, Fornerod M, de Haas V, Harrison CJ, Inaba H, Kaspers      79. Bhatt VR, Akhtari M, Bociek RG, Sanmann JN, Yuan J, Dave BJ,
    GJ, Liang DC, Locatelli F, Masetti R, Perot C, Raimondi SC,              Sanger WG, Kessinger A, Armitage JO. Allogeneic stem cell trans-
    Reinhardt K, Tomizawa D, von Neuhoff N, Zecca M, Zwaan CM,               plantation for Philadelphia chromosome-positive acute myeloid leu-
    van den Heuvel-Eibrink MM, Hasle H. t(6;9)(p22;q34)/DEK-                kemia. J Natl Compr Canc Netw. 2014;12(7):963–8.
530                                             13   Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies
80. Lazarevic VL, Labopin M, Depei W, Yakoub-Agha I, Huynh A,                   94. Patel SS, Kuo FC, Gibson CJ, Steensma DP, Soiffer RJ, Alyea
    Ljungman P, Schaap N, Cornelissen JJ, Maillard N, Pioltelli P,                  EP III, Chen YA, Fathi AT, Graubert TA, Brunner AM, Wadleigh
    Gedde-Dahl T, Lenhoff S, Houhou M, Esteve J, Mohty M, Nagler                    M, Stone RM, DeAngelo DJ, Nardi V, Hasserjian RP, Weinberg
    A. Relatively favorable outcome after allogeneic stem cell trans-               OK. High NPM1-mutant allele burden at diagnosis predicts unfa-
    plantation for BCR-ABL1-positive AML: a survey from the acute                   vorable outcomes in de novo AML. Blood. 2018;131(25):2816–
    leukemia working party of the European Society for blood and                    25. https://doi.org/10.1182/blood-2018-01-828467. Epub 2018
    marrow transplantation (EBMT). Am J Hematol. 2018;93(1):31–9.                   May 3.
    https://doi.org/10.1002/ajh.24928. Epub 2017.                               95. Perry M, Bertoli S, Rocher C, Hayette S, Ducastelle S, Barraco
81. Neuendorff NR, Burmeister T, Dörken B, Westermann J. BCR-                      F, Labussière-Wallet H, Salles G, Recher C, Thomas X, Paubelle
    ABL-positive acute myeloid leukemia: a new entity? Analysis of                 E. FLT3-TKD mutations associated with NPM1 mutations define
    clinical and molecular features. Ann Hematol. 2016;95(8):1211–21.               a favorable-risk group in patients with acute myeloid leuke-
    https://doi.org/10.1007/s00277-016-2721-z. Epub 2016 Jun 14.                    mia. Clin Lymphoma Myeloma Leuk. 2018;18:e545–50. pii:
    PMID: 27297971.                                                                 S2152-2650(18)30224-6.
82. Neuendorff NR, Schwarz M, Hemmati P, Türkmen S, Bommer C,                   96. van de Kerkhof D, Scharnhorst V, Huysentruyt CJ, Brands-
    Burmeister T, Dörken B, le Coutre P, Arnold R, Westermann J. BCR-               Nijenhuis AV, Ermens AA. Charcot-Leyden crystals in acute
    ABL1(+) acute myeloid leukemia: clonal selection of a BCR-ABL1(-)               myeloid leukemia. Int J Lab Hematol. 2015;37(4):e100–2. https://
    subclone as a cause of refractory disease with nilotinib treatment. Acta        doi.org/10.1111/ijlh.12336. Epub 2015 Mar 9.
    Haematol. 2015;133(2):237–41. https://doi.org/10.1159/000368176.            97. Taylor G1, Ivey A, Milner B, Grimwade D, Culligan D. Acute
    Epub 2014 Nov 12.PMID: 25401297 Oct 31.                                         myeloid leukaemia with mutated NPM1 presenting with exten-
83. Falini B, Mecucci C, Tiacci E, Alcalay M, Rosati R, Pasqualucci L,              sive bone marrow necrosis and Charcot-Leyden crystals. Int J
    La Starza R, Diverio D, Colombo E, Santucci A, Bigerna B, Pacini                Hematol. 2013;98(3):267–8. https://doi.org/10.1007/s12185-013-
    R, Pucciarini A, Liso A, Vignetti M, Fazi P, Meani N, Pettirossi V,             1394-9. Epub 2013 Jul 25.
    Saglio G, Mandelli F, Lo-Coco F, Pelicci PG, Martelli MF, GIMEMA            98. Wouters BJ, Löwenberg B, Erpelinck-Verschueren CA, van Putten
    Acute Leukemia Working Party. N Engl J Med. 2005;352(3):254–                    WL, Valk PJ, Delwel R. Double CEBPA mutations, but not single
    66. Erratum in: N Engl J Med. 2005 Feb 7;352(7):740.                            CEBPA mutations, define a subgroup of acute myeloid leukemia
84. Falini B, Martelli MP, Bolli N, Sportoletti P, Liso A, Tiacci E,                with a distinctive gene expression profile that is uniquely associ-
    Haferlach T. Acute myeloid leukemia with mutated nucleophosmin                  ated with a favorable outcome. Blood. 2009;113(13):3088–91.
    (NPM1): is it a distinct entity? Blood. 2011;117(4):1109–20. Epub           99. Dufour A, Schneider F, Metzeler K, et al. Acute myeloid leukemia
    2010 Oct 28.                                                                    with biallelic CEBPA gene mutations and normal karyotype rep-
85. Heath EM, Chan SM, Minden MD, Murphy T, Shlush LI, Schimmer                     resents a distinct genetic entity associated with a favorable clinical
    AD. Biological and clinical consequences of NPM1 mutations in                   outcome. J Clin Oncol. 2010;28(4):570–7.
    AML. Leukemia. 2017;31(4):798–807. https://doi.org/10.1038/                100. Mannelli F, Ponziani V, Bencini S, Bonetti MI, Benelli M, Cutini
    leu.2017.30. Epub 2017 Jan 23. Review.                                          I, Gianfaldoni G, Scappini B, Pancani F, Piccini M, Rondelli T,
86. Falini B, Nicoletti I, Martelli MF, Mecucci C. Acute myeloid leuke-             Caporale R, Gelli AM, Peruzzi B, Chiarini M, Borlenghi E, Spinelli
    mia carrying cytoplasmic/mutated nucleophosmin (NPMc+ AML):                     O, Giupponi D, Zanghì P, Bassan R, Rambaldi A, Rossi G, Bosi
    biologic and clinical features. Blood. 2007;109(3):874–85.                      A. CEBPA-double-mutated acute myeloid leukemia displays a
87. Falini B, Macijewski K, Weiss T, Bacher U, Schnittger S, Kern W,                unique phenotypic profile: a reliable screening method and insight
    Kohlmann A, Klein HU, Vignetti M, Piciocchi A, et al. Multilineage              into biological features. Haematologica. 2017;102(3):529–40.
    dysplasia has no impact on biologic, clinicopathologic, and prog-          101. Greif PA, Dufour A, Konstandin NP, Ksienzyk B, Zellmeier E,
    nostic features of aml with mutated nucleophosmin (NPM1).                       Tizazu B, Sturm J, Benthaus T, Herold T, Yaghmaie M, Dörge
    Blood. 2010;115:3776–86. https://doi.org/10.1182/blood-2009-08-                 P, Hopfner KP, Hauser A, Graf A, Krebs S, Blum H, Kakadia
    240457. [PubMed] [Cross Ref].                                                   PM, Schneider S, Hoster E, Schneider F, Stanulla M, Braess
88. Schnittger S, Bacher U, Kern W, Alpermann T, Haferlach C,                       J, Sauerland MC, Berdel WE, Büchner T, Woermann BJ,
    Haferlach T. Prognostic impact of FLT3-ITD load in NPM1 mutated                 Hiddemann W, Spiekermann K, Bohlander SK. GATA2 zinc
    acute myeloid leukemia. Leukemia. 2011;25:1297–304. https://doi.                finger 1 mutations associated with biallelic CEBPA mutations
    org/10.1038/leu.2011.97.                                                        define a unique genetic entity of acute myeloid leukemia. Blood.
89. Bennett JM, Pryor J, Laughlin TS, Rothberg PG, Burack WR. Is                    2012;120(2):395–403.
    the association of “cup-like” nuclei with mutation of the NPM1             102. Gaidzik VI, Teleanu V, Papaemmanuil E, Weber D, Paschka P,
    gene in acute myeloid leukemia clinically useful? Am J Clin Pathol.             Hahn J, Wallrabenstein T, Kolbinger B, Köhne CH, Horst HA,
    2010;134(4):648–52.                                                             Brossart P, Held G, Kündgen A, Ringhoffer M, Götze K, Rummel
90. Park BG, Chi HS, Jang S, Park CJ, Kim DY, Lee JH, Lee JH,                       M, Gerstung M, Campbell P, Kraus JM, Kestler HA, Thol F,
    Lee KH. Association of cup-like nuclei in blasts with FLT3 and                  Heuser M, Schlegelberger B, Ganser A, Bullinger L, Schlenk RF,
    NPM1 mutations in acute myeloid leukemia. Ann Hematol.                          Döhner K, Döhner H. UNX1 mutations in acute myeloid leuke-
    2013;92(4):451–7.                                                               mia are associated with distinct clinico-pathologic and genetic
91. Kunchala P, Kuravi S, Jensen R, McGuirk J, Balusu R. When the                   features. Leukemia. 2016;30(11):2282.
    good go bad: mutant NPM1 in acute myeloid leukemia. Blood Rev.             103. You E, Cho YU, Jang S, Seo EJ, Lee JH, Lee JH, Lee KH, Koh
    2018;32(3):167–83.         https://doi.org/10.1016/j.blre.2017.11.001.          KN, Im HJ, Seo JJ, Park YM, Lee JK, Park CJ. Frequency and
    Epub 2017 Nov 4. Review.                                                        clinicopathologic features of RUNX1 mutations in patients with
92. Haferlach C, Mecucci C, Schnittger S, Kohlmann A, Mancini M,                    acute myeloid leukemia not otherwise specified. Am J Clin Pathol.
    Cuneo A, Testoni N, Rege-Cambrin G, Santucci A, Vignetti M, et al.              2017;148(1):64–72.
    AML with mutated NPM1 carrying a normal or aberrant karyotype              104. Gaidzik VI, Bullinger L, Schlenk RF, Zimmermann AS, Röck J,
    show overlapping biologic, pathologic, immunophenotypic, and                    Paschka P, Corbacioglu A, Krauter J, Schlegelberger B, Ganser A,
    prognostic features. Blood. 2009;114(14):3024–32. Epub 2009                     Späth D, Kündgen A, Schmidt-Wolf IG, Götze K, Nachbaur D,
    May 8.                                                                          Pfreundschuh M, Horst HA, Döhner H, Döhner K. RUNX1 muta-
93. Medinger M, Passweg JR. Acute myeloid leukaemia genomics. Br J                  tions in acute myeloid leukemia: results from a comprehensive
    Haematol. 2017;179(4):530–42. https://doi.org/10.1111/bjh.14823.                genetic and clinical analysis from the AML study group. J Clin
    Epub 2017 Jun 27. Review.                                                       Oncol. 2011;29(10):1364–72.
13   Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies                                                         531
105. Behrens K, Triviai I, Schwieger M, Tekin N, Alawi M, Spohn M,              tic syndromes and acute myeloid leukemia by next generation
     Indenbirken D, Ziegler M, Müller U, Alexander WS, Stocking                 sequencing, a comparison with de novo diseases. Leuk Res.
     C. Runx1 downregulates stem cell and megakaryocytic tran-                  2015;39(3):348–54.
     scription programs that support niche interactions. Blood.          120.   Pantic M, Pfeifer D, Kapp-Schwoerer S, Ihorst G, Becker H,
     2016;127(26):3369–81.                                                      Zeiser R, Duyster J, Schmitt-Graeff A. TP53 abnormalities and
106. Daneshbod Y, Kohan L, Taghadosi V, Weinberg OK, Arber DA.                  chromosomal aneuploidy in acute panmyelosis with myelofibro-
     Prognostic Significance of Complex Karyotypes in Acute Myeloid             sis. Leukemia. 201933(12):2956–2962. https://doi.org/10.1038/
     Leukemia. Curr Treat Options Oncol. 2019;20(2):15. https://doi.            s41375-019-0523-8. Epub 2019 Jul 26.
     org/10.1007/s11864-019-0612-y. PMID: 30741367 Review.               121.   Bennett JM, Catovsky D, Daniel MT, et al. Proposed revised crite-
107. Seymour JF, Döhner H, Butrym A, Wierzbowska A, Selleslag D,                ria for the classification of acute myeloid leukemia. A report of the
     Jang JH, Kumar R, Cavenagh J, Schuh AC, Candoni A, Récher                  French-America n-British Cooperative Group. Ann Intern Med.
     C, Sandhu I, Del Castillo TB, Al-Ali HK, Falantes J, Stone                 1985;103:620–5.
     RM, Minden MD, Weaver J, Songer S, Beach CL, Dombret                122.   Canaani J, Beohou E, Labopin M, Socié G, Huynh A, Volin L,
     H. Azacitidine improves clinical outcomes in older patients with           Cornelissen J, Milpied N, Gedde-Dahl T, Deconinck E, Fegueux
     acute myeloid leukaemia with myelodysplasia-related changes                N, Blaise D, Mohty M, Nagler A. Impact of FAB classification
     compared with conventional care regimens. BMC Cancer.                      on predicting outcome in acute myeloid leukemia, not otherwise
     2017;17(1):852.                                                            specified, patients undergoing allogeneic stem cell transplanta-
108. Weinberg O, Gibson CJ, Blonquist TM, Neuberg D, Pozdnyakova                tion in CR1: an analysis of 1690 patients from the acute leukemia
     O, Kuo F, Ebert BL, Hasserjian RP. Association of mutations with           working party of EBMT. Am J Hematol. 2017;92(4):344–50.
     morphological dysplasia in de novo acute myeloid leukemia with-     123.   Rose D, Haferlach T, Schnittger S, Perglerová K, Kern W,
     out 2016 WHO Classification-defined cytogenetic abnormalities.             Haferlach C. Subtype-specific patterns of molecular mutations in
     Haematologica. 2018;103(4):626–63.                                         acute myeloid leukemia. Leukemia. 2017;31(1):11–7.
109. Weinberg OK, Pozdnyakova O, Campigotto F, et al. Reproducibility    124.   Bennett JM, Catovsky D, Daniel MT, et al. Proposal for the rec-
     and prognostic significance of morphologic dysplasia in de novo            ognition of minimally differentiated acute myeloid leukaemia
     acute myeloid leukemia. Mod Pathol. 2015;28(7):965–76.                     (AML-MO). Br J Haematol. 1991;78:325–9.
110. Vardiman J, Reichard K. Acute myeloid leukemia with                 125.   Arber DA. Revisiting erythroleukemia. Curr Opin Hematol.
     myelodysplasia- related changes. Am J Clin Pathol.                        2017;24(2):146–51.
     2015;144(1):29–43.                                                  126.   Wang SA, Patel KP, Pozdnyakova O, Peng J, Zuo Z, Dal Cin P,
111. Porwit A, Vardiman JW. Acute myeloid leukemia with expanded                Steensma DP, Hasserjian RP. Acute erythroid leukemia with
     erythropoiesis. Haematologica. 2011;96(9):1241–3.                          <20% bone marrow blasts is clinically and biologically similar
112. Wang SA, Hasserjian RP. Acute erythroleukemias, acute mega-                to myelodysplastic syndrome with excess blasts. Mod Pathol.
     karyoblastic leukemias, and reactive mimics. Am J Clin Pathol.             2016;29(10):1221–31.
     2015;144:44–60.                                                     127.   Calvo X, Arenillas L, Luño E, Senent L, Arnan M, Ramos F,
113. Miesner M, Haferlach C, Bacher U, Weiss T, Macijewski K,                   Ardanaz MT, Pedro C, Tormo M, Montoro J, et al. Erythroleukemia
     Kohlmann A, Klein HU, Dugas M, Kern W, Schnittger S, et al.                shares biological features and outcome with myelodysplastic
     Multilineage dysplasia (MLD) in acute myeloid leukemia (AML)               syndromes with excess blasts: a rationale for its inclusion into
     correlates with MDS-related cytogenetic abnormalities and a prior          future classifications of myelodysplastic syndromes. Mod Pathol.
     history of MDS or MDS/MPN but has no independent prognos-                  2016;29(12):1541–51.
     tic relevance: a comparison of 408 cases classified as “AML not     128.   Alkharabsheh O, Al-Kali A, Saadeh S, He R, Viswanatha D,
     otherwise specified” (AML-NOS) or “AML with myelodysplasia-               Greipp P, Reichard K, Shah M, Gangat N, Patnaik M, Hogan W,
     related changes” (AML-MRC). Blood. 2010;116(15):2742–51.                   Litzow M, Alkhateeb H, Nguyen PL. The clinical outcomes of
     Epub 2010 Jun 25.                                                          reclassified erythroleukemia (erythroid/myeloid) as myelodys-
114. Weinberg OK, Hasserjian RP, Li B, Pozdnyakova O. Assessment                plastic syndrome (MDS) per 2017 WHO guideline compared to
     of myeloid and monocytic dysplasia by flow cytometry in de novo            MDS. Am J Hematol. 2018; https://doi.org/10.1002/ajh.25239.
     AML helps define an AML with myelodysplasia-related changes                [Epub ahead of print].
     category. J Clin Pathol. 2017;70(2):109–15.                         129.   Qiu S, Jiang E, Wei H, Lin D, Zhang G, Wei S, Zhou C, Liu K,
115. Devillier R, Mansat-De Mas V, Gelsi-Boyer V, Demur C, Murati A,            Wang Y, Liu B, et al. An analysis of 97 previously diagnosed
     Corre J, Prebet T, Bertoli S, Brecqueville M, Arnoulet C, Recher           de novo adult acute erythroid leukemia patients following the
     C, Vey N, Mozziconacci MJ, Delabesse E, Birnbaum D. Role of                2016 revision to World Health Organization classification. BMC
     ASXL1 and TP53 mutations in the molecular classification and               Cancer. 2017;17(1):534. Epub 2017 Aug 9.
     prognosis of acute myeloid leukemias with myelodysplasia-          130.   Reinig EF, Greipp PT, Chiu A, Howard MT, Reichard KK. De
     related changes. Oncotarget. 2015;6(10):8388–96.                           novo pure erythroid leukemia: refining the clinicopathologic
116. Claerhout H, Lierman E, Michaux L, Verhoef G, Boeckx N. A                  and cytogenetic characteristics of a rare entity. Mod Pathol.
     monocentric retrospective study of 138 therapy-related myeloid             2018;31(5):705–17.
     neoplasms. Ann Hematol. 2018; https://doi.org/10.1007/s00277-      131.   Valent P, Sotlar K, Blatt K, Hartmann K, Reiter A, Sadovnik
     018-3462-y. [Epub ahead of print].                                         I, Sperr WR, Bettelheim P, Akin C, Bauer K, George TI,
117. Ganser A, Heuser M. Therapy-related myeloid neoplasms. Curr                Hadzijusufovic E, Wolf D, Gotlib J, Mahon FX, Metcalfe DD,
     Opin Hematol. 2017;24(2):152–8.                                            Horny HP, Arock M. Proposed diagnostic criteria and classifi-
118. Cleven AH, Nardi V, Ok CY, Goswami M, Dal Cin P, Zheng Z,                  cation of basophilic leukemias and related disorders. Leukemia.
     Iafrate AJ, Abdul Hamid MA, Wang SA, Hasserjian RP. High                   2017;31(4):788–97.
     p53 protein expression in therapy-related myeloid neoplasms is      132.   Pileri SA, Ascani S, Cox MC, Campidelli C, Bacci F, Piccioli M,
     associated with adverse karyotype and poor outcome. Mod Pathol.            Piccaluga PP, Agostinelli C, Asioli S, Novero D, Bisceglia M,
     2015;28(4):552–63.                                                         Ponzoni M, Gentile A, Rinaldi P, Franco V, Vincelli D, Pileri A
119. Ok CY, Patel KP, Garcia-Manero G, Routbort MJ, Fu B, Tang                  Jr, Gasbarra R, Falini B, Zinzani PL, Baccarani M. Myeloid sar-
     G, Goswami M, Singh R, Kanagal-Shamanna R, Pierce SA,                      coma: clinico-pathologic, phenotypic and cytogenetic analysis of
     et al. Mutational profiling of therapy-related myelodysplas-               92 adult patients. Leukemia. 2007;21:340–50.
532                                          13   Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies
133. Campidelli C, Agostinelli C, Stitson R, Pileri SA. Myeloid sar-       146. Hsu AP, Johnson KD, Falcone EL, Sanalkumar R, Sanchez L,
     coma: extramedullary manifestation of myeloid disorders. Am J              Hickstein DD, Cuellar-Rodriguez J, Lemieux JE, Zerbe CS,
     Clin Pathol. 2009;132(3):426–37.                                           Bresnick EH, Holland SM. GATA2 haploinsufficiency caused by
134. Almond LM, Charalampakis M, Ford SJ, Gourevitch D, Desai                   mutations in a conserved intronic element leads to MonoMAC
     A. Myeloid sarcoma: presentation, diagnosis, and treatment. Clin           syndrome. Blood. 2013;121(19):3830–7, S1–7.
     Lymphoma Myeloma Leuk. 2017;17:263–7.                                 147. Spinner MA, Sanchez LA, Hsu AP, Shaw PA, Zerbe CS, Calvo KR,
135. Falini B, Lenze D, Hasserjian R, Coupland S, Jaehne D, Soupir              Arthur DC, Gu W, Gould CM, Brewer CC, Cowen EW, Freeman
     C, Liso A, Martelli MP, Bolli N, Bacci F, Pettirossi V, Santucci A,        AF, Olivier KN, Uzel G, Zelazny AM, Daub JR, Spalding CD,
     Martelli MF, Pileri S, Stein H. Cytoplasmic mutated nucleophos-            Claypool RJ, Giri NK, Alter BP, Mace EM, Orange JS, Cuellar-
     min (NPM) defines the molecular status of a significant fraction of        Rodriguez J, Hickstein DD, Holland SM. GATA2 deficiency: a
     myeloid sarcomas. Leukemia. 2007;21(7):1566–70.                            protean disorder of hematopoiesis, lymphatics, and immunity.
136. Choi M, Jeon YK, Sun CH, Yun HS, Hong J, Shin DY, Kim I, Yoon              Blood. 2014;123(6):809–21.
     SS, Koh Y. RTK-RAS pathway mutation is enriched in myeloid            148. McReynolds LJ, Yang Y, Yuen Wong H, Tang J, Zhang Y, Mulé
     sarcoma. Blood Cancer J. 2018;8(5):43.                                     MP, Daub J, Palmer C, Foruraghi L, Liu Q, Zhu J, Wang W, West
137. Peterson LC, Bloomfield CD, Niemeyer CM, Döhner H, Godley                  RR, Yohe ME, Hsu AP, Hickstein DD, Townsley DM, Holland
     LA. Myeloid neoplasms with germline predisposition. In:                    SM, Calvo KR, Hourigan CS. MDS-associated mutations in
     Swerdlow SH, Campo E, Harris NL, Jaffe ES, Pileri SA, Stein H,             germline GATA2 mutated patients with hematologic manifesta-
     Thiele J, editors. WHO classification of tumours of haematopoi-            tions. Leuk Res. 2019;76:70–5.
     etic and lymphoid tissues. 4th ed. Lyon: International Agency for     149. Hasle H. Myelodysplastic and myeloproliferative disorders
     Research on Cancer; 2017. p. 122–8.                                        of childhood. Hematology Am Soc Hematol Educ Program.
138. Vinh DC, Patel SY, Uzel G, Anderson VL, Freeman AF, Olivier KN,            2016;2016(1):598–604.
     Spalding C, Hughes S, Pittaluga S, Raffeld M, Sorbara LR, Elloumi     150. Satgé D, Seidel MG. The pattern of malignancies in Down syn-
     HZ, Kuhns DB, Turner ML, Cowen EW, Fink D, Long-Priel D,                  drome and its potential context with the immune system. Front
     Hsu AP, Ding L, Paulson ML, Whitney AR, Sampaio EP, Frucht                 Immunol. 2018;(9):3058.
     DM, DeLeo FR, Holland SM. Autosomal dominant and sporadic             151. Hasle H, Clemmensen IH, Mikkelsen M. Risks of leukaemia
     monocytopenia with susceptibility to mycobacteria, fungi, papil-           and solid tumours in individuals with Down’s syndrome. Lancet.
     lomaviruses, and myelodysplasia. Blood. 2010;115(8):1519–29.               2000;355:165–9.
139. Rojek K, Nickels E, Neistadt B, Marquez R, Wickrema A, Artz           152. Bhatnagar N, Nizery L, Tunstall O, Vyas P, Roberts I. Transient
     A, van Besien K, Larson RA, Lee MK, Segal JP, King MC, Walsh               abnormal myelopoiesis and AML in Down syndrome: an update.
     T, Shimamura A, Keel SB, Churpek JE, Godley LA. Identifying                Curr Hematol Malig Rep. 2016;11(5):333–41.
     inherited and acquired genetic factors involved in poor stem cell     153. Roberts I, Izraeli S. Haematopoietic development and leukaemia
     mobilization and donor-derived malignancy. Biol Blood Marrow               in Down syndrome. Br J Haematol. 2014;167(5):587–99.
     Transplant. 2016;22(11):2100–3.                                       154. Watanabe K. Recent advances in understanding transient abnor-
140. Churpek JE. Familial myelodysplastic syndrome/acute myeloid                mal myelopoiesis in Down syndrome. Pediatr Int. 2018; https://
     leukemia. Best Pract Res Clin Haematol. https://doi.org/10.1016/j.         doi.org/10.1111/ped.13776. [Epub ahead of print].
     beha.2017.10.002.                                                     155. Saida S. Evolution of myeloid leukemia in children with Down
141. Wlodarski MW, Niemeyer CM. Introduction: genetic syn-                      syndrome. Int J Hematol. 2016;103(4):365–72. https://doi.
     dromes predisposing to myeloid neoplasia. Semin Hematol.                   org/10.1007/s12185-016-1959-5. Epub 2016 Feb 24.
     2017;54(2):57–9.                                                      156. Facchetti F, Petrella T, Pileri SA. Blastic plasmacytoid den-
142. Wlodarski MW, Hirabayashi S, Pastor V, Starý J, Hasle H,                   dritic cell neoplasm. In: Swerdlow SH, Campo E, Harris NL,
     Masetti R, Dworzak M, Schmugge M, van den Heuvel-Eibrink                   Jaffe ES, Pileri SA, Stein H, Thiele J, editors. WHO classifica-
     M, Ussowicz M, De Moerloose B, Catala A, Smith OP, Sedlacek                tion of tumours of haematopoietic and lymphoid tissues. 4th
     P, Lankester AC, Zecca M, Bordon V, Matthes-Martin S,                      ed. Lyon: International Agency for Research on Cancer; 2017.
     Abrahamsson J, Kühl JS, Sykora KW, Albert MH, Przychodzien                 p. 174–7.
     B, Maciejewski JP, Schwarz S, Göhring G, Schlegelberger B,            157. Facchetti F, Cigognetti M, Fisogni S, Rossi G, Lonardi S, Vermi
     Cseh A, Noellke P, Yoshimi A, Locatelli F, Baumann I, Strahm B,            W. Neoplasms derived from plasmacytoid dendritic cells. Mod
     Niemeyer CM, EWOG-MDS. Prevalence, clinical characteristics,               Pathol. 2016;29(2):98–111.
     and prognosis of GATA2-related myelodysplastic syndromes in           158. Suzuki Y, Kato S, Kohno K, Satou A, Eladl AE, Asano
     children and adolescents. Blood. 2016;127(11):1387–97.                     N, Kono M, Kato Y, Taniwaki M, Akiyama M, Nakamura
143. McReynolds LJ, Calvo KR, Holland SM. Germline GATA2 muta-                  S. Clinicopathological analysis of 46 cases with CD4+ and/or
     tion and bone marrow failure. Hematol Oncol Clin North Am.                 CD56+ immature haematolymphoid malignancy: reappraisal
     2018;32(4):713–28.                                                         of blastic plasmacytoid dendritic cell and related neoplasms.
144. Drazer MW, Kadri S, Sukhanova M, Patil SA, West AH, Feurstein              Histopathology. 2017;71(6):972–84.
     S, Calderon DA, Jones MF, Weipert CM, Daugherty CK, Ceballos-        159. Sullivan JM, Rizzieri DA. Treatment of blastic plasmacytoid
     López AA, Raca G, Lingen MW, Li Z, Segal JP, Churpek JE,                   dendritic cell neoplasm. Hematology Am Soc Hematol Educ
     Godley LA. Prognostic tumor sequencing panels frequently iden-             Program. 2016;(1):16–23.
     tify germ line variants associated with hereditary hematopoietic      160. Khoury JD. Blastic plasmacytoid dendritic cell neoplasm. Curr
     malignancies. Blood Adv. 2018;2(2):146–50.                                 Hematol Malig Rep. 2018;13(6):477–83.
145. Dickinson RE, Griffin H, Bigley V, Reynard LN, Hussain R,             161. Sapienza MR, Abate F, Melle F, Orecchioni S, Fuligni F, Etebari
     Haniffa M, Lakey JH, Rahman T, Wang XN, McGovern N, Pagan                  M, Tabanelli V, Laginestra MA, Pileri A, Motta G, Rossi M,
     S, Cookson S, McDonald D, Chua I, Wallis J, Cant A, Wright M,              Agostinelli C, Sabattini E, Pimpinelli N, Truni M, Falini B,
     Keavney B, Chinnery PF, Loughlin J, Hambleton S, Santibanez-              Cerroni L, Talarico G, Piccioni R, Amente S, Indio V, Tarantino
     Koref M, Collin M. Exome sequencing identifies GATA-2 muta-                G, Brundu F, Paulli M, Berti E, Facchetti F, Dellino GI, Bertolini
     tion as the cause of dendritic cell, monocyte, B and NK lymphoid           F, Tripodo C, Rabadan R, Pileri SA. Blastic plasmacytoid den-
     deficiency. Blood. 2011;118(10):2656–8.                                    dritic cell neoplasm: genomics mark epigenetic dysregulation as
13     Acute Leukemia of Myeloid, Lymphoid, and Ambiguous Lineage and Related Malignancies                                                         533
       a primary therapeutic target. Haematologica. 2018; https://doi.              Characterization      of      Leukemias     (EGIL).       Leukemia.
       org/10.3324/haematol.2018.202093. pii: haematol.2018.202093.                 1995;9(10):1783–6.
       [Epub ahead of print].                                                  172. Zhou Y, Fan X, Routbort M, Cameron Yin C, Singh R, Bueso-
162.   Tang Z, Li Y, Wang W, Yin CC, Tang G, Aung PP, Hu S, Lu X,                   Ramos C, Thomas DA, Milton DR, Medeiros LJ, Lin P. Absence
       Toruner GA, Medeiros LJ, Khoury JD. Genomic aberrations                      of terminal deoxynucleotidyl transferase expression identifies a
       involving 12p/ETV6 are highly prevalent in blastic plasmacytoid              subset of high-risk adult T-lymphoblastic leukemia/lymphoma.
       dendritic cell neoplasms and might represent early clonal events.            Mod Pathol. 2013;26(10):1338–45.
       Leuk Res. 2018;73:86–94.                                                173. Zheng W, Medeiros LJ, Young KH, Goswami M, Powers L,
163.   Borowitz MJ, Béné MC, Harris NL, Porwit A, Matutes E, Arber                  Kantarjian HH, Thomas DA, Cortes JE, Wang SA. CD30 expres-
       DA. Acute leukemias of ambiguous lineage. In: Swerdlow SH,                   sion in acute lymphoblastic leukemia as assessed by flow cytom-
       Campo E, Harris NL, Jaffe ES, Pileri SA, Stein H, Thiele J, edi-             etry analysis. Leuk Lymphoma. 2014;55(3):624–7.
       tors. WHO classification of tumours of haematopoietic and lym-          174. Jain N, Lamb AV, O’Brien S, Ravandi F, Konopleva M, Jabbour
       phoid tissues. 4th ed. Lyon: International Agency for Research on            E, Zuo Z, Jorgensen J, Lin P, Pierce S, Thomas D, Rytting M,
       Cancer; 2017. p. 180–7.                                                      Borthakur G, Kadia T, Cortes J, Kantarjian HM, Khoury JD. Early
164.   Heesch S, Neumann M, Schwartz S, Bartram I, Schlee C,                        T-cell precursor acute lymphoblastic leukemia/lymphoma (ETP-
       Burmeister T, Hänel M, Ganser A, Heuser M, Wendtner CM, Berdel               ALL/LBL) in adolescents and adults: a high-risk subtype. Blood.
       WE, Gökbuget N, Hoelzer D, Hofmann WK, Thiel E, Baldus                       2016;127(15):1863–9.
       CD. Acute leukemias of ambiguous lineage in adults: molecular           175. Wang P, Peng X, Deng X, Gao L, Zhang X, Feng Y. Diagnostic
       and clinical characterization. Ann Hematol. 2013;92(6):747–58.               challenges in T-lymphoblastic lymphoma, early T-cell
165.   Béné MC. Biphenotypic, bilineal, ambiguous or mixed lineage:                 precursor acute lymphoblastic leukemia or mixed pheno-
       strange leukemias! Haematologica. 2009;94(7):891–3.                          type acute leukemia. A case report. Medicine (Baltimore).
166.   Béné MC, Porwit A. Acute leukemias of ambiguous lineage.                     2018;97(41):e12743.
       Semin Diagn Pathol. 2012;29(1):12–8. Review.                            176. Wenzinger C, Williams E, Gru AA. Updates in the pathology of
167.   Porwit A, Béné MC. Acute leukemias of ambiguous origin. Am J                 precursor lymphoid neoplasms in the revised fourth edition of the
       Clin Pathol. 2015;144(3):361–76.                                             WHO classification of tumors of hematopoietic and lymphoid tis-
168.   Lao ZT, Ding LW, An O, Hattori N, Sun QY, Tan KT, Mayakonda                  sues. Curr Hematol Malig Rep. 2018;13(4):275–88.
       A, Chuan WG, Madan V, Lin DC, Yang H, Koeffler HP. Mutational           177. Swerdlow SH, Campo E, Pileri SA, Harris NL, Stein H, Siebert R,
       and transcriptomic profiling of acute leukemia of ambiguous lineage          Advani R, Ghielmini M, Salles GA, Zelenetz AD, Jaffe ES. The
       reveals obscure but clinically important lineage bias. Haematologica.        2016 revision of the World Health Organization classification of
       2018; https://doi.org/10.3324/haematol.2018.202911. pii: haema-              lymphoid neoplasms. Blood. 2016;127(20):2375–90.
       tol.2018.202911. [Epub ahead of print].                                 178. Borowitz JM, Chan JKC, Downing JR, Le Beau MM, Arber
169.   Borowitz MJ, Chan JKC, Béné M-C, Arber DA. T-lymphoblastic                   DA. B-lymphoblastic leukemia/lymphoma, not otherwise speci-
       leukemia/lymphoma. In: Swerdlow SH, Campo E, Harris NL,                      fied. B-lymphoblastic leukemia/lymphoma with recurrent genetic
       Jaffe ES, Pileri SA, Stein H, Thiele J, editors. WHO classifi-               abnormalities. In: Swerdlow SH, Campo E, Harris NL, Jaffe ES,
       cation of tumours of haematopoietic and lymphoid tissues. 4th                Pileri SA, Stein H, Thiele J, editors. WHO classification of tumours
       ed. Lyon: International Agency for Research on Cancer; 2017.                 of haematopoietic and lymphoid tissues. 4th ed. International
       p. 209–12.                                                                   Agency for Research on Cancer: Lyon; 2017. p. 200–9.
170.   Belver L, Ferrando A. The genetics and mechanisms of T cell acute       179. Pui CH, Roberts KG, Yang JJ, Mullighan CG. Philadelphia
       lymphoblastic leukaemia. Nat Rev Cancer. 2016;16(8):494–507.                 chromosome-like acute lymphoblastic leukemia. Clin Lymphoma
171.   Bene MC, Castoldi G, Knapp W, Ludwig WD, Matutes E, Orfao                    Myeloma Leuk. 2017;17(8):464–70.
       A, van’t Veer MB. Proposals for the immunological classifica-           180. Tasian SK, Loh ML, Hunger SP. Philadelphia chromosome-like
       tion of acute leukemias. European Group for the Immunological                acute lymphoblastic leukemia. Blood. 2017;130(19):2064–72.