Thankamma V Ajithkumar MD FRCP FRCR, Helen Hatcher PHD MRCP - Specialist Training in Oncology, 1e-Mosby (2011)
Thankamma V Ajithkumar MD FRCP FRCR, Helen Hatcher PHD MRCP - Specialist Training in Oncology, 1e-Mosby (2011)
This book is dedicated to our mothers (Thankamma and Margaret) who gave us life
and the encouragement to write, and to our families for keeping us sane during the
preparation of this book.
Edinburgh London New York Oxford Philadelphia St Louis Sydney Toronto 2011
© 2011 Elsevier Ltd. All rights reserved.
No part of this publication may be reproduced or transmitted in any form or by any means, electronic or
mechanical, including photocopying, recording, or any information storage and retrieval system, without
permission in writing from the publisher. Details on how to seek permission, further information about the
Publisher’s permissions policies and our arrangements with organizations such as the Copyright Clearance Center
and the Copyright Licensing Agency, can be found at our website: www.elsevier.com/permissions.
This book and the individual contributions contained in it are protected under copyright by the Publisher (other
than as may be noted herein).
Notices
Knowledge and best practice in this field are constantly changing. As new research and experience broaden our
understanding, changes in research methods, professional practices, or medical treatment may become necessary.
Practitioners and researchers must always rely on their own experience and knowledge in evaluating and using
any information, methods, compounds, or experiments described herein. In using such information or methods
they should be mindful of their own safety and the safety of others, including parties for whom they have a
professional responsibility.
With respect to any drug or pharmaceutical products identified, readers are advised to check the most current
information provided (i) on procedures featured or (ii) by the manufacturer of each product to be administered,
to verify the recommended dose or formula, the method and duration of administration, and contraindications.
It is the responsibility of practitioners, relying on their own experience and knowledge of their patients, to make
diagnoses, to determine dosages and the best treatment for each individual patient, and to take all appropriate
safety precautions.
To the fullest extent of the law, neither the Publisher nor the authors, contributors, or editors, assume any
liability for any injury and/or damage to persons or property as a matter of products liability, negligence or
otherwise, or from any use or operation of any methods, products, instructions, or ideas contained in the
material herein.
The
Working together to grow publisher’s
policy is to use
libraries in developing countries paper manufactured
from sustainable forests
www.elsevier.com | www.bookaid.org | www.sabre.org
Printed in China
Contributors
ix
Preface
It is a startling fact that one in three people have tried to explain the practical elements of
will develop cancer in their lifetime and one in treating cancer patients from radiotherapy plan-
four people will die of cancer. It is therefore ning to chemotherapy prescription, the rationale
important to most people in the health sector to behind them and what the patients are likely to
have a basic understanding of cancer and its ask about these treatments.
treatments. Oncology is one of the most rapidly changing
When we started our training in oncology and areas of medicine with the developments of new
were faced with new patients every day, what we therapies constantly evolving. With the modern
felt was needed was a book which summarized computer-literate patient, we are repeatedly chal-
the most common cancers and their treatments. lenged to be up to date, which is difficult in such
There are many books which discuss the scien- a dynamic specialty. In some instances, we have
tific basis of cancer, but we wanted to know what included website addresses, which are useful
to tell the patient about what treatments they either for your patients or yourselves.
may have and what their outcome might be. This In essence, this is a handbook of practical
is true for many health professionals, both those guidance and knowledge for all those who come
in training (including students) and those whose into contact with patients going through their
main speciality is not cancer management, but cancer journey. It is the culmination of years of
which brings them into contact with cancer teaching and our own knowledge journey, which
patients regularly, e.g. general practitioners, we hope provides the reader with rapid access to
specialist nurses and pharmacists. essential information.
We wanted to provide an overview of clinical
presentation, required investigations and treat- TV Ajithkumar and
ment options according to stage and fitness. We HM Hatcher
x
Acknowledgements
We are indebted to those who took time in their • Dr Evis Sala, Cambridge University Hospital,
busy lives to provide their expert comments on Cambridge.
the manuscript: • Varian Medical Systems.
• Professor Ann Barrett We thank Sue Shadbolt for her excellent organi-
• Professor Ian Judson zational and computer skills in assisting with the
• Dr Mike Shere preparation of figures.
• Dr Sharon Straus We are grateful to Timothy Horne, Janice
Urquhart and Fiona Conn at Elsevier for com-
We owe thanks to several of our colleagues for
missioning this book and supporting us through-
the provision of figures and illustrations:
out its development and production.
• Dr Matt Beasley, Dr Priyanka Mehta, Neil Finally, we thank all those who helped us
Ogborne, Henry Lawrence, Alison Stapleton through our training and the knowledge from
and Helen Appleby at Bristol Oncology senior colleagues to students whose questions led
Centre. us to search deeper.
• Dr Andrew Medford at Glenfield Hospital.
• Mr Rob Grimer at Royal Orthopaedic Hos-
pital, Birmingham.
xi
Clinical approach to
cancer patients
TV Ajithkumar and HM Hatcher
1
Introduction Investigations for
Cancer is a global healthcare problem. In a suspected cancer
the year 2000, cancer accounted for 12% of Initial investigations are to establish a histologi-
approximately 56 million deaths worldwide cal diagnosis of cancer, to assess the extent of
from all causes. It is estimated by 2050 more local disease, and to look for metastatic disease.
than 27 million cancer cases per year will be Further investigations are done to evaluate suit-
diagnosed and result in 17.5 million deaths. In ability for standard or trial treatment and to
the UK, 1 in 3 adults will develop some form of assess the severity of any co-morbid medical
cancer during their lifetime and 1 in 4 die from conditions.
cancer.
A suspicion of cancer or diagnosis of cancer is Diagnostic investigations
the beginning of a difficult journey. In many Diagnostic investigations are undertaken in a
ways it is like climbing a mountain for the first logical order, with blood tests including relevant
time; a journey filled with uncertainty and chal- tumour markers and simple imaging, and proceed
lenges which carries no guarantees (Figure 1.1). to the diagnostic imaging of choice for particular
Patients often carry additional anxieties if there tumour types. Imaging helps to determine local
have been perceived delays so that a knowledge and distant metastatic staging. Confirmation
of warning signs of cancer and an understanding of the diagnosis can be obtained by cytology
of referral and treatment is essential for any or biopsy. Many tumours require biopsy for
healthcare professional.
Patients presenting to their primary care physi-
cian with warning signs of cancer (Box 1.1)
Box 1.1: Warning signs of cancer
should be referred to a specialist centre for an
• Rapid weight loss
urgent evaluation. In the UK and many parts of
• Haemoptysis/persistent cough/hoarseness of voice
the World, cancer treatment is organized around for >3 weeks
a multidisciplinary team which consists of physi- • Rectal bleeding/melaena/altered bowel habit
cians, surgeons, cancer specialists, radiologists, • Haematuria
pathologists and clinical nurse specialists. Many • Breast lump
investigations for suspected cancer are done • Progressive dysphagia
either in ‘one-stop’ (e.g. for breast cancer) or • Persistent headache
‘two-stop’ clinics (e.g. lung cancer) to expedite • Persistent non-specific symptoms
diagnosis and treatment.
Figure 1.1
The diagnosis of cancer can be a lonely and challenging
experience. Figure 1.3
CT scan abdomen shows bilateral adrenal metastasis in
small cell lung cancer.
Don’ts
• Don’t give life expectancy in fixed terms. Though many patients on palliative treatment ask about their life
expectancy, it is better to refrain from giving some answers like 4–5 weeks or less than 6 months. It is always a
good idea to ask patient whether they would like to get some figure or just a rough idea. It is better is put in
terms like ‘weeks to months’ or ‘months to years’. If you are forced to give a figure, give a broad range and
explain that the figure is an average. For example, for some with lung cancer with disseminated liver metastasis
the estimated life expectancy will be around 3–6 months, but explain that in the best case scenario it might be
more than 6 months and in the worst case less than 3 months.
• Don’t be too authoritative.
• Don’t try to answer a question if you don’t know the right answer.
• Don’t give equivocal results or bad news over phone – you don’t know what is going on the other end of the
phone or what support the patient has. If you have good news, just let the patient know as soon as possible.
Anxiety for the patient is often greatest when awaiting a scan result. Some centres have a policy of
telephoning with good results, but beware that an intelligent patient will know when there is bad news if a
result is not given (patients talk to each other as well as to professionals).
of the information overload can add to the morbidities at the diagnosis, likely responsive-
anxiety. A clinician role is to help the patient to ness of the particular tumour type and calculating
cope with the situation, and give them clear direc- the tolerance to any planned treatment.
tions on further management and aim (Box 1.3). Performance status (PS): Performance status
helps to quantify the physical well-being of
patients and helps to determine optimal treat-
Assessing fitness for treatment ment, make treatment modifications (including
Assessing fitness for treatment is an important dose modification of chemotherapy) and to
part of decision making. It is mainly based on measure the intensity of supportive care required.
the performance status of patient, active co- It should be clearly documented at the beginning
6
CLINICAL APPROACH TO CANCER PATIENTS 1
and throughout treatment. There are a number small and potentially curable. Similarly, many
of scoring systems and the two most commonly chemotherapy drugs have systemic organ effects
used are Karnofsky performance status (KPS) which need to be taken into account when plan-
and WHO score (Table 1.1). Patients are gener- ning anticancer treatment.
ally eligible for curative treatment only if PS
is 0–1, palliative anticancer treatment when PS Implementing planned
is 0–2 and generally no anticancer treatment is
given if PS 3–4. However, in certain situations, treatment
when the disease is very responsive to treatment For many early stage cancers, delay in treatment
and rapid deterioration is due to the current is proven to compromise the chances of cure (e.g.
disease process, modified curative treatment germ cell tumours), and in tumours with rapid
is considered (e.g. germ cell tumours). If per- proliferation (e.g. Burkitt’s lymphoma which has
formance status deteriorates rapidly during anti- a doubling time of <24 hours) even a few day of
cancer treatment, treatment is either stopped or delay in initiating treatment can be detrimental.
modified. Hence it is important to start curative treatment
A proper assessment of active co-morbidities as soon as possible.
is important in predicting side effects to proposed
cancer treatment. For example, patients with
severe COPD with poor pulmonary function
Follow-up during treatment
tests (FEV1 <1) are not considered for surgery or Follow-up during treatment is to assess response
radical radiotherapy even if the tumour is very to treatment, monitor toxicity and modify further
7
I BASIC PRINCIPLES OF CANCER MANAGEMENT
treatment. Toxicity should be monitored along- practitioners will have a key role in coordinating
side treatment so that patients on a 3 weekly some of this care and providing additional
cycle of chemotherapy will be seen every 3 weeks necessary services such as physiotherapy and
prior to each treatment to modify the dose or occupational therapy.
adjust supportive treatments. For patients who At the time of diagnosis, an individual should
have had significant toxicities on a previous therefore be provided with sources of informa-
cycle, they may be seen in between the cycle tion to help them through all these areas to facili-
dates. Patients undergoing radiotherapy will tate their treatment and return to health. The key
often be seen at least once throughout their treat- areas are:
ment to assess acute toxicities and prescribe sup-
• Psychosocial support.
portive measures as necessary.
• Income support.
The optimal timing to assess tumour response
• Macmillan team.
to treatment varies depending upon the type of
• Cancer care in the community.
cancer treatment. For example, radiotherapy and
hormonal agents generally take a 3–4 months to The potential sources of this support can be in
show any response to treatment, whereas chemo- the form of documents from helpful websites or
therapy response is faster (6–9 weeks). Hence, organizations, local cancer support groups, and
investigations to assess response to treatment named key workers such as specialist nurses or
should be planned according to the appropriate Macmillan nurses. It should be emphasized at the
time scale. However if there is any clinical suspi- start of diagnosis and treatment that it is natural
cion of progression while on treatment, prompt to require the help of others at some point in the
investigations are required prior to planned journey and that it will only help their overall
investigations. outcome. The special needs of children diagnosed
with cancer and their family is discussed on
p. 320.
Support during treatment
From the moment of diagnosis of cancer, a per- Cancer screening
son’s life is changed forever. It affects not only
Several cancers now have proven screening pro-
their physical health, but their mental health is
grammes which have been seen to improve either
also challenged. Different individuals deal with
the incidence or outcome of invasive cancers, e.g.
this process in different ways but each should be
breast and cervix. For other cancers screening
offered appropriate support for their needs. This
programmes are in development to try to mini-
may involve psychological support or something
mize the mortality from this disease.
more practical such as directing them to possible
sources of financial support. Many people are
working at the time of diagnosis and their future Genetic screening
employment may be affected depending on their Patients with a suspected genetic component to
job and employer. Laws exist to protect individu- their cancer need referral to a clinical genetics
als, but those who are self-employed will only be department. Chapter 5 (p. 45) deals with the
protected if they had pre-existing insurance. recommendations for referral to clinical genetics
Relationships are frequently challenged by the and further management for the patient and their
diagnosis of cancer and the journey through its family.
treatment, and it is not uncommon for couples
to split up at this time. Financial worries may Follow-up after treatment and
continue after treatment has completed as it
affects the ability to gain new life insurance or a
management of recurrence
mortgage. Those living in countries which require Almost all patients who have been treated with
health insurance will notice an increase in premi- curative intent need some regular follow-up to
ums or difficulty obtaining further health insur- detect an early potentially curable recurrence
ance. Local health providers such as general and a second cancer. The frequency and mode of
8
CLINICAL APPROACH TO CANCER PATIENTS 1
Further reading
Figure 1.5 Referral guide for suspected cancer, NICE. Available from
Successful treatment of cancer is often a cause for http://guidance.nice.org.uk/CG27/Guidance/pdf/English
celebration, but people will also need help getting
down from the mountain at the other side.
9
Principles of
surgical oncology
TV Ajithkumar
2
Introduction nodes, aspiration of cysts, diagnosis of thyroid
nodule and confirmation of recurrent or meta-
Surgery remains the major mode of cure for static disease.
many cancers. The majority of solid tumours are Core biopsy helps to visualize architecture as
treated initially with a curative resection if pos- well as to perform immunohistochemical studies.
sible. The proportion of patients who can undergo It is useful in the diagnosis of solid masses such
a curative resection depends on the stage at pres- as breast lumps or liver metastases. However,
entation and tumour site. Curative surgery may core biopsy should not be used in lymphoma
result in disruption of cosmesis and function. (which requires extensive immunohistochemical
Hence surgery also has an important role in stains) or for soft tissue or bone sarcoma.
reconstructive surgery to maintain body image Incisional biopsy is used when core biopsy is
and ability to return to normal functioning life. non-diagnostic and excision biopsy is not appro-
The potential role of surgery in the manage- priate. The common indication is a suspected
ment of cancer includes: sarcoma needing neoadjuvant treatment or defi-
• Diagnosis and staging. nite resection. Care should be taken when plan-
• Curative. ning incision biopsy to ensure that the site of
• Palliative. biopsy is within the area definite surgery and
• Reconstructive (oncoplastic) surgery. should only be undertaken by surgeons who will
• Risk reduction surgery. undertake the final surgery. A poorly planned
• Minimal access and robotic. incision biopsy can lead to unnecessary morbid
surgery.
Excision biopsy involves removal of the entire
Diagnosis and staging mass or skin lesion. It is important to make sure
that this procedure does not compromise a later
Diagnosis wider excision if necessary. The specimen needs
Histological diagnosis is obtained from tumour to be oriented in three dimensions for the pathol-
tissue by fine needle aspiration cytology, core ogist to determine surgical margins.
biopsy, incisional biopsy and excisional biopsy. Frozen section is occasionally used preopera-
Choosing the right technique is based on the tively to confirm diagnosis when previous histo-
location of tumour, anticipated type of the logic diagnosis is not available (e.g. solitary lung
tumour and reliability of the method to make a lesions), to decide need for further surgery (e.g.
definite diagnosis. lymph node dissection) and ensure adequate sur-
Fine needle aspiration (FNA) yields quick gical margins.
result and shows cellular characteristics but
not architecture. FNA serves as good screening Staging
tool prior to more definitive diagnostic methods. Various surgical procedures such as endoscopy
FNA is useful for diagnosis of enlarged lymph and staging laparotomy aid in defining the extent
10 © 2011, Elsevier Ltd
DOI: 10.1016/B978-0-7234-3458-0.00007-5
PRINCIPLES OF SURGICAL ONCOLOGY 2
of disease as well as obtaining histological con- abdominal hysterectomy with bilateral
firmation of metastatic disease. These are covered salpingo-oophorectomy).
in detail for specific malignancies. One example
At the time of surgery exposure and shedding
is the use of laparoscopy as an adjunct to detect
viable tumour cells should be avoided if possible.
small peritoneal and liver metastases reduces the
Certain tumours have a propensity to recur
number of ‘open and close’ laparotomies by less
along surgical incision lines or drainage sites e.g.
than 5% in stomach cancer.
mesothelioma and sarcoma. A curative resection
is aimed at a gross visible margin as imaging
Curative surgery modalities will not identify microscopic disease.
Surgery plays a significant role in curative treat- This gross margin depends on the type of malig-
ment of cancer. For many cancers surgery is the nancy and pattern of local spread. An adequate
primary modality of treatment. A decision regard- gross margin helps to ensure adequate micro-
ing curative surgery is made after careful consid- scopic margin of resection. Table 2.1 shows
eration of various patient and tumour related examples of the gross resection margin and
characteristics at a multidisciplinary meeting with microscopic resection margin for some common
surgeons, oncologists, radiologists and patholo- tumours.
gists. At this meeting staging and diagnosis will be Multimodality treatment has affected the
reviewed along with information concerning the surgical approach to many cancers. The use of
patient. Patient-related factors which influence the radiotherapy, chemotherapy or both has led to
choice of curative surgery include age, perform- the use of less radical procedures with an improve-
ance status and co-morbidities. Tumour-related ment of quality of life.
factors include chances of long-term benefit and
potential surgical risks and complications. Surgery of regional lymph nodes
Surgery of the primary tumour Some tumours spread to regional lymph nodes
in a predictive fashion. In these cancers, local
Curative surgery is aimed at removal of the
lymph nodes in continuity with lymphatics are
malignant tumour with a clear margin of normal
removed along with the primary tumour. This
tissue (‘R0’ resection) with reconstruction of the
provides important staging information as well
surgical defect if appropriate. Based on the extent
as a therapeutic advantage to minimize the risk
of removal of cancer, resections are classified as
of a regional recurrence, preventing more exten-
follows which is part of the TNM staging:
sive surgery in the future. Various methods are
• R0 – all margins are histologically free of used to screen pathological involvement of lymph
tumour. nodes before extensive lymph nodes dissection is
• R1 – microscopic residual disease after undertaken. These include:
resection.
• FNA of enlarged regional lymph nodes.
• R2 – gross residual disease after resection.
• Node sampling – involves cherry picking
The tumour should be orientated and marked of 4–5 regional lymph nodes (e.g. breast
at the time of surgery such that any positive cancer).
margins can be identified anatomically should • Sentinel node biopsy – is based on the princi-
the need for re-excision arise. ple that tumour spread to a single node (sen-
Depending on the type of cancer and anatomi- tinel) prior to spreading to other nodes in an
cal site, curative surgery can be: ‘ordered’ fashion and lymph nodes can be
identified by using blue dye or radioisotopes
• Wide local excision (e.g. breast cancer).
(Figure 2.1).
• Removal of part of the organ and sur
• Lymph node dissection
rounding tissue at risk of spread (e.g. partial
gastrectomy). However, in many situations the role of lymph
• Removal of an entire organ with or without node dissection in overall survival remains
important adjacent structures (e.g. total controversial. In practice, patients with involved
11
I BASIC PRINCIPLES OF CANCER MANAGEMENT
Table 2.1: Gross resection margin and minimal microscopic resection margin for curative resection
Tumour type Local spread pattern Gross margin Microscopic margin
Breast cancer Circumferential 1 cm 1–5 mm
Lung cancer Within the same lobe Lobectomy
Oesophageal cancer Longitudinal submucosal 5 cm 1 mm
Stomach cancer Submucosal and towards serosa 1 cm 1 mm
Colon cancer Submucosal and towards serosa 3 cm 1 mm
Rectal cancer Towards serosa 3 cm 1 mm
Squamous cell carcinoma Peripheral 1 cm 1 mm
skin
Malignant melanoma Deep 1–2 cm 1 mm
Head and neck cancer Within the anatomic Anatomic compartment 1 mm
compartment
A B
Figure 2.1
Sentinel node biopsy. Sentinel node is identified by either use of blue dye (A) or radioisotope (Tc-99m albumin
nanocolloid). Picture B shows an intense uptake above the injection site with smaller less distinct nodes.
lymph nodes undergo node dissection whereas disease at presentation or in a very select group
the role of elective lymph node dissection is of patients with good performance status with a
dependent on the site of cancer, type of cancer long disease-free survival after treatment of the
and other prognostic factors. Benefits of nodal primary tumour. In the absence of proper
dissections in different cancers are discussed in randomized studies it is not known whether the
the corresponding chapters. observed therapeutic benefit is actual or due to
the strict selection process (selection bias). The
common situations are resection of isolated or
Metastatectomy as part of limited liver metastases in colorectal cancer
curative surgery which results in 20–40% 5-year survival
Resection of isolated metastatic lesions is (p. 164). Pulmonary metastatectomy in sarcomas
useful for a selected group of patients in certain leads to a 5-year survival of 20–25% (p. 259).
cancers (Box 2.1). In general this is undertaken An alternative is the evolving use of radio
for patients with surgically resectable metastatic frequency ablation.
12
PRINCIPLES OF SURGICAL ONCOLOGY 2
Box 2.1: Role of metastatectomy Box 2.2: Potential indications for risk
reducing surgery
• Liver – selected patients with colorectal cancer.
• Lung – selected patients with colorectal, kidney Surgery Indication
and testicular cancers and sarcoma.
Bilateral mastectomy BRCA1/2 mutations
• Adrenal – selected patients with resectable lung Familial breast cancer
cancer and isolated adrenal disease. Unilateral breast cancer in
• Brain – solitary metastasis with controlled/ <40 years
potentially curable systemic disease. Bilateral oophorectomy BRCA1/2 mutations
Familial ovarian cancer
Total proctocolectomy Familial adenomatous
polyposis or APC
mutations
Salvage surgery
Hereditary non-polyposis
Surgery is useful as a salvage measure after colon cancer – germline
primary treatment failure or recurrence after mutations
definitive treatment. It is only appropriate for Thyroidectomy RET oncogene mutation
MEN 2
fit patients with a good chance of prolonged
survival. Examples include abdomino-perineal
resection after chemoradiotherapy for anal
cancer, and exenteration in cervical cancer after
chemoradiotherapy. In patients with prior limited
surgery or chemoradiotherapy, a second chance Risk reduction surgery
of cure is aimed with surgery. Examples include
Less than 5% patients have a genetic component
mastectomy for local recurrence after conserva-
to their cancer (p. 45). Increasing understanding
tive surgery for breast cancer and neck node
of the development of genetically associated
dissection for isolated nodal recurrence after
cancers has led to prophylactic surgery for some
chemoradiotherapy for head and neck cancer.
patients. Box 2.2 shows the indications for
common prophylactic surgeries. Appropriate
Palliative surgery genetic testing and counselling is, however, an
The aim of palliative surgery is to improve or absolute pre-requisite prior to any prophylactic
prevent significant symptoms (e.g. pain, bleeding surgery. Women with BRCA1 and BRCA2 muta-
and obstruction) which are likely to occur tions have a high risk of breast cancer which is
without intervention. It can also enhance the reduced by 90–95% with bilateral mastectomy.
effect of chemotherapy (e.g. in ovarian cancer However, the decision to undergo prophylactic
where it improves survival). Debulking surgery mastectomy should be done after careful discus-
of the primary tumour can produce good sion on explaining the future quality of life,
symptom control and improve quality of life by potential surgical risks and wishes of the patient.
preventing complications due to uncontrolled Alternative risk reduction methods such as use of
disease (e.g. loco-regional surgery in breast tamoxifen and prophylactic oophorectomy after
cancer prevents fungation and symptomatic axil- completion of family should also be considered.
lary disease). Another example is FAP and prophylactic colec-
tomy (p. 50).
Reconstructive (oncoplastic)
surgery Minimal access and
Extensive resection often results in disruption of
robotic surgery
normal anatomy and subsequent cosmesis and The role of minimal access surgery is being
function. Plastic surgical techniques are useful in increasingly studied in the management of solid
correcting the anatomical defects and improving tumours. It is being more often used in abdomi-
cosmesis (e.g. reconstructive breast surgery) and nal malignancies and studies show that laparo-
function (e.g. in head and neck surgery). scopic surgery results in the same long term
13
I BASIC PRINCIPLES OF CANCER MANAGEMENT
survival as that of open surgery, with no increased Robotic assisted surgery is being studied in
risk of abdominal wall recurrence, less surgical prostate and renal cell cancers. This technique
morbidity and quick return to normal function. may result in minimal surgical trauma with better
Laparoscopic surgery is an accepted modality toxicity profile.
of surgical treatment in gastric, kidney, adrenal
and colorectal cancers. Video-assisted thoracic Further reading
surgery for stage I lung cancer is a particularly Sabel M, Sandak V and Sussman J. Essentials of surgical
attractive option for elderly patients. Oncology. 2006, Elsevier.
14
Principles of radiotherapy
TV Ajithkumar
3
Introduction apy given either alone or in combination with
chemotherapy or biological agents) or adjuvant
After surgery, radiotherapy is the most effective radiotherapy (given after definitive treatment of
curative treatment for cancer, contributing up to tumour, usually surgery). Palliative radiotherapy
25–30% of cure. At least half of the patients with is intended to improve symptoms (e.g. bone pain)
cancer require radiotherapy at some time in their or prevent tumour related complications (e.g.
illness of which about 60% are treated with cura- ulceration in breast cancer).
tive intent, often in combination with surgery
and chemotherapy. Radiotherapy involves use of
various types of ionizing radiation, and X-ray is Methods of delivery
the commonest type of radiation used. Other of radiotherapy
forms of radiation include electrons, protons,
neutrons and gamma radiations from radioactive Radiotherapy is either delivered by a radiation
isotopes. This chapter intends to review the prin- source placed away from the body (teletherapy
ciples of practical radiotherapy (Box 3.1) and a or external beam radiotherapy), by placing a
detailed discussion on radiobiology and mathe- radiation source into the tumour (interstitial
matical modelling are beyond the scope of this brachytherapy, e.g. small tongue cancer) or in a
chapter. body cavity containing the tumour (intracavitary
brachytherapy, e.g. cervical cancer) or by intra-
venous or oral administration of nonsealed
Indications for radiotherapy radionuclides. Depending on the site and type of
cancer, radiotherapy is delivered by either one of
Radiotherapy is indicated if it cures or improves
these techniques or a combination. Sometimes
the chances of cure, improves local tumour
radiotherapy is delivered concurrently with
control, achieves symptom control or improves
chemotherapy or biological agents to improve
the quality of life. There are already adequate
the chances of cure (see below).
data on these various indications for radiother-
apy for different neoplasms. With current under-
standing of radiation biology and improved
External beam radiotherapy (EBRT)
techniques, more indications are evolving, par- EBRT commonly utilizes X-rays and electrons.
ticularly for tumours previously thought to be Before the 1950s, radiotherapy units were kilo-
radioresistant (e.g. liver and pancreatic cancer). voltage machines which produced X-rays with
limited penetrability. These machines, which are
still used in some centres, can be one of the
Intent of radiotherapy following:
Radiotherapy treatment is given with either • Superficial X-rays – operate at a potential of
curative or palliative intent. Curative treatment 50–150 kVp and tube current of 5–10 mA
involves either radical radiotherapy (radiother- and useful to treat superficial skin cancers.
Box 3.1: Steps in practical radiotherapy Box 3.2: Linear accelerator (Figure 3.1)
1. Indication for radiotherapy Electrons from gun are accelerated before they hit
2. Intent of radiotherapy the target. The X-rays produced from the target are
collimated by primary collimators in the direction of
3. Which is the best way to deliver radiotherapy? patient. This beam is further modified by flattening
• External beam radiotherapy alone (e.g. head filter, secondary collimation and multileaf collimators
and neck cancer) before reaching patient’s surface.
• Brachytherapy alone (e.g. early stage cervical
cancer)
• External beam radiotherapy and brachytherapy
(e.g. advanced stage cervical cancer)
• External beam radiotherapy with concurrent Modern radiotherapy is based on megavoltage
chemotherapy (e.g. squamous oesophageal X-rays (photons) and electrons. Megavoltage
cancer)
X-rays are produced by artificial acceleration of
• External beam radiotherapy with target agents
(e.g. advanced head and neck cancer) electrons through a vacuum to impact on a target
• Radioisotopes (e.g. well-differentiated thyroid in machines called linear accelerators (LINACs)
cancer) (Box 3.2). The energy of the X-rays is propor-
4. Obtaining informed consent tional to the speed of electrons. Electrons are
• Benefit of radiotherapy produced when the target in a linear accelerator
• Explaining the process is removed from the path of the electron beam
• Expected side effects – short-term and (Box 3.2). Modern LINACs have facilities to
long-term
produce both photons and electrons. Electrons
5. Radiotherapy planning and delivery
have a predictable penetrability and hence are
• Position of patient and immobilization
useful when it is important to limit the radiation
• Localization of tumour (CT or MRI)
dose to a deeper organ. Characteristics of photons
• Target volume definition
that are useful in their clinical use are:
• Tumour volumes (GTV, CTV, PTV)
• Normal organs • Greater penetration compared to lower energy
• Planning technique X-rays, which helps in delivering a higher
• Conventional (2D) proportion of dose to the deep tumour com-
• Conformal planning (3D) pared to the surface dose.
• Intensity-modulated radiotherapy (IMRT) • Skin sparing effect – the maximum absorbed
• Image guided radiotherapy (IGRT) dose is deposited a few millimetres beneath
• Treatment planning the skin. This helps to deliver higher dose to
• Beam arrangement depth without causing significant damage to
• Beam size and shape skin and subcutaneous tissue (Box 3.3).
• Energy of beam
• Wedges, shields, bolus Brachytherapy
• Evaluation of treatment plans – homogenous Brachytherapy involves placing radioactive
dose to PTV and dose to critical organs sources at a very short distance from or in contact
• Prescription of radiotherapy treatment with, the tumour. Rapid fall off of radiation dose
• Treatment verification and corrections at increasing distance from the source permits
6. Supportive care during radiotherapy delivery of a high dose of radiotherapy to the
7. Treatment modifications during treatment (e.g. tumour with sparing of surrounding normal
bank holidays, toxicities and interruptions)
tissue. Short half life and high specific activity of
8. Care after radiotherapy and survivorship issues
isotopes allow delivery of the dose over a short
period of time. The spatial distribution of sources
• Deep (ortho – ‘orthodox’) X-rays – operate at is based on a complex set of rules.
a potential of 150–500 kVp and tube current The previous problems of radiation safety for
of 10–20 mA. These are useful to treat thick patients and staff, and prolonged treatment time,
skin cancers and rib lesions. In many centres, have become history with modern brachytherapy
these machines are being replaced by electron machines (Figure 3.3). Clinical applications of
16 beam treatment. brachytherapy are as follows:
PRINCIPLES OF RADIOTHERAPY 3
Electron
Target
Primary collimator
Multileaf collimator
Multileaf collimator
A B
Figure 3.1
A & B, Linear accelerator. (Courtesy of Varian Medical Systems.)
60
% depth dose
40
20
95%
0 90%
0 5 10 15 20 25
80%
Depth (cm) 10 MeV
6.53 cm
15 MeV 50%
18 MeV 30%
22 MeV
6 MV Beam size defined by
A 10 MV 90% isodose at depth
B
Figure 3.2
A, The percentage of depth dose of electrons (MeV) and photons (MV). B, Electron beam isodose curves – Note the
constriction of 90% isodose at depth which necessitates addition of wider margins to beam width for adequate
coverage of tumour at depth.
A B
Figure 3.3
Brachytherapy machine and its application. A, High dose rate (HDR) afterloading brachytherapy machine which is
fully computer controlled and completes treatment in a few minutes. B, Interstitial brachytherapy in breast cancer
(Courtesy of Varian Medical Systems).
over a period of time) (Box 3.5) and side effects Radiotherapy planning
(p. 348). Side effects of radiotherapy depend on
the area of treatment, total dose and dose per and delivery
fraction. These can be acute (occurring during
radiotherapy and within 3 months of completion Position and immobilization
of radiotherapy) or chronic (occurring 3 months Radiotherapy aims to deliver a uniform (homog-
after completion of radiotherapy). enous) therapeutic dose of radiation to the tumour
18
PRINCIPLES OF RADIOTHERAPY 3
Box 3.4: How does radiation work? • Direct damage causes single or double strand break
• Hydroxyl group leads to single strand break
Therapeutic use of radiotherapy is born out of
empiricism. Science caught up with empirical clinical
use later to provide explanations for various patterns X-rays γ-rays
of response to treatment. The cancer cell has an
unlimited capacity to multiply and radiotherapy is H2O
aimed to prevent this unlimited multiplication. The
conventional explanation of radiation effect is based Ionization
on radiation damage to DNA caused directly by
photons and indirectly by free radicals which are
H+ + OH•
produced when photons interact with water
molecules in the tissue (see Figure 3.4). There are
three different types of radiation damage to DNA:
• Lethal – irreversible, irreparable leading to cell
death.
• Sublethal – under normal circumstances cells can A G A A C G T
be repaired in a few hours unless additional
sublethal damage (another dose of radiation)
occurs which makes the damage lethal. T C T T G C A
• Potentially lethal – the component of radiation
damage can be modified by the post radiation
environment.
After DNA damage the following changes can occur:
However, DNA damage is not the only mechanism of • DNA repair
radiotherapy action. Radiotherapy can also influence
• Loss of unrestricted cell division and later cell death
genes controlling the cell cycle (e.g. RB gene, p53)
• Programmed cell death (apoptosis)
and alter the expression of various genes resulting in
expression of cytokines, growth factors, structural
proteins or enzymes. Radiation damage to cell
membrane sends signals to the nucleus and these
signals affect cell behaviour. Sensors
Box 3.5: Rationale for fractionation Box 3.6: Volumes in radiotherapy (Figure 3.5)
Radiotherapy usually results in equal damage to both The following are the volumes in radiotherapy:
normal cells and cancer arising from them. However, • GTV – Gross tumour volume – is the gross disease
normal cells and cancer cells differ in their ability to or radiologically visible tumour – this area has 109
recover from radiation damage. This differential cancer cells. In the figure, the T2W MRI shows
capacity of normal cells to repair and re-grow is hyperintense low grade brain tumour.
exploited in eradication of tumour cells. Radiation
sensitivity of cells is dependent on the phase of cell • CTV – Clinical target volume – is the volume
cycle they are in: G2 and M are radiosensitive which contains GTV and/or subclinical microscopic
whereas S phase is radioresistant. At the time of disease (106 cells) which need to be treated to an
radiotherapy, the cancer cells are in various phases of adequate dose to achieve the aim of treatment.
cell cycles; some of them will be in the sensitive The CTV depends on the pattern of local spread of
phase when there is a good chance of cell kill and specific tumours. In the figure, note the outer
some will be in the resistant phase. border of CTV merges with that of PTV as there is
no possibility of microscopic spread beyond the
Rationale of fractionation is explained by 5 Rs. Some inner layer of the skull.
of the Rs lead to more cancer cell kill, whereas others
lead to better repair of normal cells. • PTV – Planning target volume – is a geographical
concept which denotes the CTV and margins for
• Redistribution of cells – delivering radiotherapy as geographic variations and inaccuracies during
multiple treatments allows tumour cells to reassort treatment. The margin added to CTV to derive
into more radiosensitive phase leading to more PTV depends mainly on the immobilization device
cancer cell damage. (e.g. 2–3 mm with stereotactic frame). PTV has
• Reoxygenation – many cancers have hypoxic areas two components:
within the tumour and normal cells do not have • IM (internal margin) – is a margin added to
any hypoxic areas. Hypoxic areas are relatively CTV to compensate for normal anatomical
resistant to radiotherapy. With each fraction, movements and variations in size, shape and
radiotherapy reduces the number of cancer cells position of CTV during treatment. In the figure,
and allows some of the hypoxic areas to become there is very little anatomic movement of CTV
more oxygenated leading to better cancer cell and hence IM is almost zero, whereas in a lung
radiation damage. tumour, it may be around 1–4 cm depending
• The inherent radiosensitivity of the tumour is on the location of the tumour and depth of
important in achieving a cure or control of breathing.
tumour. Some tumours are very radiosensitive (e.g. • SM (set up margin) – a number of factors can
lymphoma and seminoma) whereas others are cause uncertainties such as daily variation in
relatively radioresistant (e.g. melanoma, sarcoma). position of the patient, mechanical
Most tumours are moderately radiosensitive. Cure uncertainties of the equipment, transfer set up
can be achieved with a smaller dose of errors etc. In brain tumour treatment, if a
radiotherapy in very radiosensitive tumours (e.g. Perspex shell is used a 5 mm margin is added.
germinoma, p. 275)
• TV – treated volume – is the volume enclosed by
• Repair of sublethal damage – reducing the dose an isodose surface, selected and specified by the
per fraction helps to repair damage of normal oncologist as being appropriate to achieve the
tissues more effectively than tumours. Normal purpose of treatment.
tissue may be able to repair sublethal damage if
the interval between two fractions of radiotherapy • IrV – irradiated volume – is that tissue volume
is at least 6 hours. However, some types of cancers which receives a dose that is considered significant
also have quick ability to repair radiation damage in relation to normal tissue tolerance.
leading to radioresistance (e.g. melanoma). • PRV – planning organ at risk volume – refers to
• Repopulation – fractionation helps rapidly the definition of margins around organs at risk for
populating normal tissues such as skin and gut to injury by radiation.
repopulate and hence recover from radiotherapy Please note, even if the actual tumour (GTV) is small,
damage. Again, cancer cells also have the capacity after adding various margins as above the irradiated
to repopulate especially when radiation treatment volume is much bigger. This is one of the limiting
is very prolonged or interrupted (gap correction, factors in delivering a high dose to tumour without
see later). having significant irradiation to normal tissue. Studies
show that a 4–5% increase in dose increases the
tumour control probability by 10% and hence newer
developments are aiming at better localization of
tumour and reduction of various margins to increase
Localization of tumour the dose to the actual tumour.
Localization of tumour is to define various treat-
ment volumes (Box 3.6). Conventional localiza-
tion (2-dimensional, 2D) is with orthogonal
X-rays using special treatment planning machines
20
PRINCIPLES OF RADIOTHERAPY 3
called simulators. These are diagnostic X-ray accuracy of GTV delineation. Planning CT scan
machines, which can mimic all the positions and is usually taken at a slice interval of 1.5–5 mm.
movements of a treatment machine (Figure 3.6A) A slice thickness of <3 mm helps to get better
and some of these machines have a CT scan facil- resolution of digitally reconstructed images
ity. 2D localization is appropriate for palliative (DRRs) which are used to check accuracy of
treatment. Radical radiotherapy utilizes a plan- treatment delivery (see later in Box 3.12).
ning CT scan or advanced techniques to define
the various tumour volumes in 3-dimensions. Planning technique
Modern computer software allows co-registra- Conventional (2D) simulator planning uses
tion of the planning CT scan with an MRI or simple beam arrangements such as single field
PET scan (Figure 3.6B), which improves the radiotherapy or parallel opposed beams. In con-
formal (3D) planning various volumes and organs
at risk (OAR) are delineated individually on
every slice of cross-sectional imaging and a treat-
ment plan produced by a planning computer.
Intensity modulated planning (IMRT) is a further
improvement of 3D planning, which is aimed at
ensuring homogeneous dose to the tumour with
less dose to organs at risk. This technique has
particular advantage with concave-shaped target
volumes with OAR located in the concavity. The
technique is also useful in sparing critical organs
from radiotherapy toxicity (e.g. parotid gland
sparing in head and neck cancer).
Image guided radiotherapy (IGRT) is a further
advance in radiotherapy planning and delivery
where the real time position of the tumour is
taken into consideration (Figure 3.7). 3D, IMRT
Figure 3.5 and IGRT use multiple beams to obtain the
Volumes in radiotherapy. optimal treatment plan.
A B
Figure 3.6
Localization of tumour. A, Simulator used for conventional localization as well as for plan verification (courtesy
Varian Medical Systems). B, Co-registered planning CT with PET scan showing primary tumour (arrow head) and PET
positive right paratracheal node (arrow).
21
I BASIC PRINCIPLES OF CANCER MANAGEMENT
A B
Figure 3.7
A&B, IMRT and IGRT. A, IMRT is a conformal technique which helps to selectively avoid/limit radiotherapy dose to
critical structures. This IMRT plan aims to give 70 Gy to ethmoid cancer (yellow line represents 70 Gy) while keeping
the dose to the optic chiasma (arrow) to less than 50 Gy. B, IGRT – specialized radiotherapy machine with on-board
imaging system (arrows) allows real time imaging and correction of error. (Courtesy Varian Medical Systems.)
Box 3.7: Radiotherapy planning (see Figure 3.8) Box 3.8: Beam shaping (Figure 3.9)
Three fields are used and the dose is prescribed at In the early 1950s radiotherapy beams used to be
the intersection point as 100% of prescribed dose either square or rectangular. There was an option to
Note the beam arrangement – shield part of the field with lead or equivalent
material. When conformal radiotherapy started
• Only one beam passes directly through the spinal evolving, individual blocks were made based on the
cord so as to keep cord dose within tolerance BEV of individual beams to shape the radiotherapy
• Beams enter only through the side of disease. No fields using cerrobend (an alloy of bismuth, lead, tin
beams are entering through the normal lung side and cadmium with a melting point of 70°C and hence
• Beams are sized according to the beam’s eye view easy to make different shapes). Currently beam
and are of different dimension shaping is achieving with MLCs (see Figure 3.9).
A C
physicists come up with more than one plan for prescribed dose. This is confirmed by looking
the oncologist to evaluate and choose one for at the dose distribution at every slice of plan
treatment. Evaluation of the plan takes the fol- or looking at the dose volume histogram
lowing steps (Box 3.9): (DVH). DVH plots the total dose against the
percentage of target volume or OAR irradi-
• Homogenous dose to the PTV – ideally, the
ated (Box 3.9). Hotspots are areas outside the
whole of the PTV should be covered by 95%
PTV which receive a dose >100% of the speci-
isodose and the recommendation is that PTV
fied PTV dose. Hotspots are considered sig-
dose should be within –5% and +7% of the
nificant only if they exceed >15 mm in
diameter, except in small organs (e.g. eye,
Box 3.9: Evaluation of a treatment plan optic nerve) where <15 mm diameter has to
(Figure 3.10)
be considered significant.
Treatment plan evaluation is to ensure that the PTV • Dose to OAR – dose to the OAR should not
receives a homogenous dose and the doses to the
OAR are within tolerance. Spatial distribution displays exceed their tolerance dose. Tolerance dose
ensure that 95% isodose encloses the PTV (Figure (TD) is the dose beyond which there is a risk
3.10A & B). Dose volume histogram (Figure 3.10C) is of dose limiting toxicity. TD is stated as TD5/5
helpful to indicate underdosage to PTV as well as
excessive dose to OAR. (dose at which there is 5% risk of occurring
dose limiting toxicity at 5 years) and TD5/50
PTV
95% isodose
(red shade)
B
A
100
90 PTV receives >95% dose,
80 there are areas receiving
>105% dose, there is no PTV
70 receiving underdosage
Volume (%)
60
50 Maximum dose to spinal cord
40 is less than 40% of
prescribed dose
30
20
10
0
0 20 40 60 80 100
Table 3.1: Normal tissue radiation tolerance dose (treated with 2 Gy per fraction)
TD5/5 (Gy)
Organ Whole 2/3rd 1/3rd Dose limiting toxicity
Brain 45 50 60 Necrosis
Brain stem 50 53 60 Necrosis
Colon 45 – 55 Obstruction, perforation and ulceration
Heart 40 45 60 Pericarditis
Kidney 23 30 50 Renal failure
Lens 10 – – Cataract
Liver 30 35 50 Hepatitis
Lung 17.5 30 45 Pulmonary fibrosis
Oesophagus 55 58 60 Stricture
Optic apparatus 50 – – Blindness
Parotid 32 32 – Xerostomia
Rectum 60 – – Necrosis, fistula, stenosis
Retina 45 – – Blindness
Spinal cord 47 50 50 Myelitis
Small intestine 40 – 50 Obstruction, perforation
Stomach 50 55 60 Ulceration, perforation
(dose at which there is 50% risk of occurring The prescription should indicate the total
dose limiting toxicity at 5 years. The aim of dose, number of fractions, dose per fraction, and
treatment plan evaluation is to limit the dose duration of treatment (e.g. 50 Gy in 20 fractions
to OAR below TD5/5. Table 3.1 gives the over 4 weeks). Conventional fractionation is to
approximate TD of various organs. deliver one radiotherapy fraction per day for 5
• Dose to other normal tissue – it is important days a week. There are a number of alternate
to check that there is no increased dose to the fractionation regimes (Box 3.10). The total dose
entry and exit of the beam and there are no of radiotherapy depends on the type of cancer,
sensitive structures in the path of the radia- site and bulk of disease (Box 3.11). Gray (Gy) is
tion beams. the usual measure of radiotherapy which is
defined as 1 joule of energy per 1 kg of material.
Dose prescription for The subunit is centigray (cGy) which is 1/100 of
radiation treatment a Gray. Radiotherapy alone for cure needs a dose
The prescription point of radiotherapy depends of >60–65 Gy as 1.8–2 Gy per day whereas adju-
on the type of plan. For a single beam, the prescrip- vant radiotherapy uses 50–60 Gy. Most studies
tion point either lies in the point of maximum dose indicate a direct relation between the dose and
(dmax) or at a depth. For parallel opposed beams chances of control and cure.
the prescription point is mid-plane whereas for all
3D and 4D planning the prescription point is often Treatment verification and corrections
at the isocentre or at the intersection of points. Accurate position and immobilization during
For electrons, dose is prescribed at 100%; planning and treatment are important in accurate
however, it is necessary that 90% isodose covers delivery of radiotherapy. In spite of this, treat-
the PTV. ment delivered can be different from the planned 25
I BASIC PRINCIPLES OF CANCER MANAGEMENT
Box 3.10: Fractionation schedules Box 3.11: Tumour control probability (TCP),
normal tissue complication probability (NTCP)
• Conventional fractionation – delivers 1.8–2 Gy
and therapeutic ratio (Figure 3.11)
single fraction per day, 5 times weekly.
• Accelerated fractionation – aims to shorten the For many types of cancer, a higher dose of
treatment time by delivering larger than standard radiotherapy produces better tumour control. After
size fractions five times weekly or more than five fractionated radiotherapy, the total number of
fractions per week of 2 Gy. Multiple fractions may cancer cells surviving depends on the initial number
also be given daily. The acute toxicity is greater of cells and the proportion of cancer cells killed with
with this and late toxicity is either the same as (if each treatment. Studies show that visible disease
complete repair of sublethal damage occurs) or needs more dose than microscopic disease (to
greater than conventional fractionation. It is pathologically detect microscopic disease, cell
useful for tumours with a rapid growth rate. aggregates of ≥106/cm3 are needed) and subclinical
disease (cannot be detected microscopically). For
• Hyperfractionation – aims to improve tumour example a dose of >65 Gy is needed to control visible
control probability with the same late effects as epithelial tumours, whereas the dose needed to
conventional fractionation. It delivers a larger control microscopic disease is 60–65 Gy and 50–60 Gy
number of smaller than conventional dose for subclinical disease.
fractions per day and the total dose is 10–20%
higher than standard fractionation while total The aim of radiotherapy is to deliver a dose of
treatment time remains the same. It is useful for radiation to destroy the tumour without leading to
tumours with slow growth. serious normal tissue complications. Therapeutic ratio
is used to represent this concept where it is the ratio
• Accelerated hyperfractionation – combines the of the tumour control probability (TCP) and normal
principles of hyperfractionation and accelerated tissue complication probability (NTCP) at a specified
fractionation. level of response (usually <5% or 0.05) for normal
• Continuous accelerated hyperfractionation tissue.
(CHART) – treatment is given as 1.5 Gy per The probability of tumour control without normal
fraction, three times daily 6 hours apart for 12 tissue complications is highest in the therapeutic
continuous days. Proved to improve local control window. The further the NTCP curve to the right of
in lung cancer. the TCP (see Figure 3.11) cure it is easier to achieve a
• Hypofractionation – delivers more than 2 Gy per tumour control with minimal toxicity (A) and a larger
fractions and less than five fractions per week. therapeutic ratio. When NTCP and TCP (C) get closer
the therapeutic ratio gets smaller.
In practical radiotherapy a better therapeutic ratio
treatment. Hence it is important to ensure that can be achieved by:
treatment delivery is within the tolerance limit • Advanced treatment planning (3D, IMRT or IGRT)
of accepted variation from the planning. The • Accurate target localization which helps to reduce
safety margins
accepted tolerance generally depends on the
• Sophisticated dose delivery
immobilization method; e.g. a variation of
<5 mm is acceptable with Perspex. The accuracy
of planned treatment can be verified by:
Box 3.12: Verification of treatment (Figure 3.12)
• Verification of treatment position before DRR (Figure 3.12A) is used to verify treatment
treatment using simulator – the simulator accuracy. This is commonly done by comparison of
X-rays are compared with DRRs for accuracy DRR with the treatment verification film (Figure
3.12B). The treatment set-up is checked by comparing
in terms of isocentre and comparison of bony the position of isocentre, bony landmarks and the
landmarks if feasible and comparison of indi- positions of MLCs or beam shaping block for the
vidual fields (Box 3.12). same beams. If a systemic error is found during
verification, it needs to be corrected before
• Verification of treatment during treatment proceeding with further treatment and the corrected
with portal imaging – a comparison is made treatment plan should be verified for accuracy of
with DRR to assess the position of isocentre treatment set-up.
in at least two views (anterior and posterior),
position of beam in relation to bony or other
visible landmarks (Box 3.12) and shape of During the process of verification two types of
individual fields. errors in treatment set up can occur:
• Real time verification of treatment with cor- • Systemic Σ – error between planning and
rection for variation is the back bone of image treatment. This needs correction before pro-
guided radiotherapy. ceeding with further treatment.
26
PRINCIPLES OF RADIOTHERAPY 3
100 100
‘Radiosensitive’ tumour
90 A TCP 90 Standard tumour
80 C NTCP 80 ‘Radioresistant’ tumour
Tumour control without
70 70 normal tissue complications
Tumour control (%)
Complications (%)
Normal tissue complication
60 60
50 50
40 40
30 30
20 20
10 10
Accepted complication rate
0
0 10 20 30 40 50 60 70 80 90 100
Dose (Gy)
Figure 3.11
TCP and NTCP (see Box 3.11).
Figure 3.12
A&B, Verification of
treatment.
B
A
• Random ∂ – variations and uncertainties that and check blood counts. Studies have shown that
arise between each fraction of treatment. It is during radical radiotherapy of head and neck,
difficult to correct this error as this does not lung, oesophageal and cervical cancer, it is
occur consistently. important to maintain a haemoglobin level of
During determination of PTV, these errors are >12 gm/dl (a level lower than this may result in
taken into consideration to add a margin to PTV. 10–12% less chance of disease control). The
The suggested margin is 2.5 Σ + 0.7∂. importance of level of haemoglobin is less clear
in other cancers. However, symptomatic anaemia
Supportive care during radiotherapy needs to be corrected. It is also important to
It is important to follow up patients regularly make sure that the blood counts remains safe (i.e.
during radiotherapy to assess and manage radio- platelet count of >100 × 109/dL and neutrophil
therapy toxicities, to monitor nutritional status count of >1.5 × 109/dL) during radiotherapy
27
I BASIC PRINCIPLES OF CANCER MANAGEMENT
especially when it is given concurrently with In the long term, there are a number of side
chemotherapy or when a significant amount of effects needing special attention. Patients receiv-
bone marrow bearing area is being irradiated. ing radiotherapy to whole or part of the brain
Blood counts below the safe threshold necessitate are at risk of cognitive impairment, cerebrovas-
either withholding chemotherapy or suspending cular accidents, hormonal deficiency and second
chemotherapy until the blood count recovers. cancer (p. 58, late effects). Radiotherapy to the
chest is associated with increased risk of second
Treatment modifications cancer and radiation damage to heart, particu-
larly in patients receiving radiotherapy for left
during treatment
breast cancer. Pelvic radiotherapy often results in
Gap correction menopause and infertility in women and sexual
One of the common situations is an interruption dysfunction and infertility in men. Radiotherapy
during radiotherapy due to holidays, non-attend- induced or promoted second cancer is an impor-
ance or intolerable toxicity. This might need tant survivorship issue. After cancer treatment,
modification of treatment. Studies have shown people often face an array of challenges including
that tumours exhibit accelerated repopulation psycho-social functioning and economic well-
during radiotherapy. Accelerated repopulation being. As physicians, our triumph is not just in
implies an increase in multiplication of cancer curing or controlling cancer, but also in helping
cells which often starts with partial shrinkage cancer survivors to lead a high quality life in the
of tumour after radiotherapy or chemotherapy. society. For a detailed discussion on cancer sur-
Prolonged interruption of radiotherapy results in vivorship, see p. 58.
decreasing efficacy of radiotherapy due to accel-
erated repopulation. Using modelling analysis, an Advances in radiotherapy
increase of total treatment duration by 1 day may
reduce local control by 1.4%. Hence attempts Based on experimental studies, there is a direct
should be made to avoid prolongation of overall correlation between the radiation dose and
treatment. Still, occasionally treatment gets pro- chances of control of cancer, though not linear.
longed, when adjustment should be made to com- However, various uncertainties in the radiother-
plete the treatment in the planned overall time by: apy planning process often lead to a large treat-
ment volume (see Box 3.6). Advances in
• delivering twice daily fractions, at a minimum radiotherapy are aimed at understanding and
of a 6-hour interval controlling these uncertainties in radiotherapy
• treating over the weekend planning. By doing so, we will be able to give
• use of biologically equivalent dose in fewer a much higher dose to the tumour and areas
fractions to achieve planned overall time. of possible spread with minimal dose to the
If treatment still cannot be completed within surrounding normal organs. The areas under
the planned overall time additional fractions may research include advanced immobilization,
be considered. A rough estimate is an additional improved radiotherapy with image guidance and
dose of 0.6 Gy per every day of increase in incorporation of molecular imaging for tumour
overall treatment time. volume delineation.
29
Principles of systemic
therapy
HM Hatcher
4
Introduction Chemotherapy
Systemic therapies form the basis for medical Chemotherapy involves the treatment with cyto-
oncology and a significant amount of clinical toxic chemicals to kill cancer cells. Its benefit
oncology in the UK. The description is broad and depends upon the high proliferation rate of
can be thought to include any treatment which cancer cells compared with the non cancer cells
is given to a patient which will be absorbed sys- in the body. The discovery of nitrogen mustard
temically and could potentially affect the whole and its effects on proliferating cells was discov-
body. Traditionally this would be thought to be ered in both World Wars, but only put into prac-
chemotherapy but the definition also includes tice to treat leukaemias and lymphomas in the
hormonal treatments, chemical adjuncts, immu- 1940s (Figure 4.1). Later that decade aminop-
notherapy, biological agents and systemic radio- terin was shown to induce remissions in leukae-
therapy treatments. Each will be discussed and mia, although the majority of patients relapsed
examples given with reference to the appropriate and died. In the following 10 years different
chapters for more detail. classes of chemotherapeutic agents were discov-
This chapter gives an overview of the practical ered. Most of these were directed against DNA
aspects of various systemic therapies (Box 4.1). synthesis or cell division. In the 1970s cytotoxic
A detailed pharmacological discussion is beyond agents were seen to impact significantly on the
the scope of this book but further reading is sug- cure of specific cancers such as testicular cancer
gested for those interested. and leukaemia (Box 4.5). This improvement in
survival was due not only to the development of
Aim of systemic treatment new drugs but also to the understanding of how
Before recommending or prescribing a systemic to combine them.
treatment the aim of the treatment has to be
understood (Box 4.2). This, in addition to a
knowledge of the specific disease and treatments Classes of chemotherapy drugs
that are effective for that tumour, will dictate the Chemotherapy agents are divided into different
type of treatment offered and its likely intensity categories according to their mechanism of
(Box 4.3). Treatment intensity should be greatest action. The rationale behind chemotherapy is
in those conditions where the intention is cure, to inhibit or kill rapidly dividing cancer cells.
and there is some evidence in certain tumours This may be due to the drug acting at a par-
(e.g. bone tumours) that increased toxicity during ticular point in the cell cycle (cell cycle-specific)
chemotherapy is related to improved survival. or is independent of the cell cycle (cell cycle-
When the treatment is not curative significant non specific) (Figure 4.2). Boxes 4.6 and 4.7
toxicity is unacceptable. give examples of different classes of drugs in
In acute leukaemias the curative chemother- each of these categories with some specific
apy is given in various phases (Box 4.4). examples.
30 © 2011, Elsevier Ltd
DOI: 10.1016/B978-0-7234-3458-0.00009-9
PRINCIPLES OF SYSTEMIC THERAPY 4
Box 4.1: Types of systemic treatments Box 4.4: Phases of chemotherapy treatment in
• Chemotherapy (single agent or in combination) haematologic malignancies (p. 310)
• Targeted treatments: Induction
• hormonal treatments e.g. tamoxifen, arimidex, Initial intense chemotherapy designed to rapidly
goserelin reduce tumour burden and aim for remission.
• antibodies e.g. rituximab, cetuximab Consolidation
• small molecules e.g. imatinib, erlotinib Continued treatment to reduce the tumour burden
further to the point of remission where no tumour
• Other agents:
cells are detectable.
• bisphosphonates, e.g. pamidronate, zoledronic
acid Maintenance
• immunological agents, e.g. interleukin-2, Ongoing lower dose treatment to maintain remission.
interferon Evidence from ALL studies show improved survival
with prolonged low-dose chemotherapy and
intrathecal methotrexate (p. 310).
Box 4.5: Examples of tumours in which Box 4.6: Cell cycle specific chemotherapy drugs
chemotherapy has a significant impact
Antimetabolites S phase e.g.
Cured with chemotherapy
methotraxate,
• Childhood acute leukaemia capecitabine
• Testicular cancer Vinca alkaloids M phase e.g. vincristine,
• Ovarian germ cell tumours vinorelbine
Taxanes M phase e.g. paclitaxel,
• Choriocarcinoma
docetaxel
• Wilms’ tumour Epipodophyllotoxins G2, S, e.g. etoposide
premitotic,
Associated with improved survival
topo II
• Breast cancer Camptothecans S phase, e.g. irinotecan,
• Osteosarcoma topo I topotecan
• Ewing’s sarcoma
• Ovarian cancer
• And several others
S
(2–6h) G2 Vinca alkaloids
(2–32h)
tumours receive additional courses of chemother-
M Mitotic inhibitors
(0.5–2h) apy to bring down the number of tumour cells to
an absolute minimum. However this does not
Taxoids necessarily result in complete removal of tumour
Alkylating agents
cells at the end of chemotherapy and it is believed
that the normal body immunosurveillance will
G1 help to achieve a cure in some instances. In some
(2-∞h) patients, cancer can grow back at any point of
time (relapse) during the conventionally undetec-
G0 table phase (see Box 4.8). In some other patients,
Figure 4.2 the tumours do not respond to chemotherapy
Cell cycle showing phases of cell replication and division. (resistance to treatment) which requires change
Cancer cells progress rapidly through this but may be of treatment (Figure 4.3).
inhibited at various points.
Rationale for combining
chemotherapy agents
chemotherapy regime is given to a sensitive In clinical practice cancer cells tend to develop
tumour it causes cell death in a proportion of the resistance to a single drug by further gene muta-
cancer cells (log kill). In a chemo-sensitive tumour, tions, or development of cellular pumps which
each course of chemotherapy (see Box 4.9) results reduce the dose of drug received by the tumour
in a proportional cell kill and with a few courses cells. Consequently the tumour will have a period
of chemotherapy, tumour may not be detectable of sensitivity followed by a rebound tumour
by conventional means (called complete response regrowth. This may be partly due to the fact that
to treatment). At this point there is a possibility not all tumour cells are passing through the same
of <109 cells remaining and stopping treatment at point of the cell cycle at the same time. Combin-
this point may lead to an early progression/relapse ing drugs allows the oncologist to direct agents
of disease. Hence patients with chemo-sensitive with different activities or against different parts
32
PRINCIPLES OF SYSTEMIC THERAPY 4
Chemo
109
Detectable cancer
No of cancer cells
(tumour burden
of 109 cells equal to
Chemo 1 gm or 1 cm tumour)
Cancer not
detectable by
conventional means
(less than 109 cells)
Carcinogenesis Cure
Point of clinical diagnosis
Time 0 Start Rx 3 weeks 6 weeks 9 weeks 12 weeks
Figure 4.3
Cancer growth and response to treatment.
Box 4.8: Why not give treatment more often to With G-CSF No G-CSF
prevent tumour regrowth?
Neutrophils (x109/L)
Box 4.9: Cycles of chemotherapy Box 4.10: How do you decide which drugs to
combine for a given cancer?
What is a cycle?
A cycle is a complete treatment from the first day of • Activity and scheduling: Each drug must have
one cycle to the first day of the second cycle. For shown activity for that type of cancer when given
many regimens this means a 3 or 4 week period of as a single agent and be given in a similar
time (depending on the dose determining toxicity scheduling and dosing (synergistic activity).
and its recovery period). Over that time drugs may all • Mechanism of action: Ideally the drugs should
be given on day 1 only or can be given at several have different, complementary mechanisms of
time points in those few weeks. action (a measure to prevent resistance).
How many cycles of chemotherapy? • Toxicities: The toxicities of the drugs should also
be different to minimize overall toxicity.
For curative treatment, further cycles of
chemotherapy are needed after a conventional e.g. Combination of gemcitabine and carboplatin in
clinical response, to tackle the clinically undetectable lung cancer:
tumour burden (Figure 4.3). Hence the amount of Activities and scheduling
chemotherapy may vary from a few cycles to months
• Carboplatin and gemticabine – each has a single
to years depending on the specific cancer (e.g. acute
agent response of ≤20% in lung cancer and is
lymphatic leukaemia needs more than a year of
given 3-weekly.
chemotherapy, whereas in choriocarcinoma two more
courses of chemotherapy is given after a complete Mechanism of action (Figure 4.5)
tumour marker response). • Carboplatin (cell cycle non-specific) act by forming
For many adjuvant treatments, traditionally 6 cycles DNA adducts which prevents DNA synthesis
of chemotherapy are given. (e.g. breast cancer • Gemcitabine (cell cycle specific) – pyramidine
usually 6–8 courses of chemotherapy depending on antimetabolite
the regime).
In palliative treatments it is important to assess Toxicity
response (usually after 2–3 courses of chemotherapy) • Carboplatin: dose limiting toxicity is
before continuing with treatment but if responding thrombocypenia
patients may continue for up to 6 (to 8) cycles • Gemcitabine: dose limiting toxicity is neutropenia
depending on toxicity and the drugs involved. Combination of carboplatin and gemcitabine results
The concept of a cycle does not fit with certain in 30–50% response rate and the dose limiting
therapies such as hormonal treatments or with toxicity is thrombocytopenia.
tyrosine kinase inhibitors as these drugs are
continued for a finite number of years or until
progression.
Gemcitabine
S
folates essential to the synthesis of nucleic acids (2–6h)
G2
and DNA (S phase) preventing the correct incor-
poration of purines or pyrimidines in the DNA M
structure. They therefore prevent DNA replica-
tion and lead to cell death. They are most effec-
tive against tumours with a high growth fraction Carboplatin
and similarly have predominant toxicities on
normal tissues with high cell turn over (e.g. G1
mucosal epithelium and gastrointestinal tract).
Examples are methotrexate, cytosine arabino-
side, capecitabine, gemcitabine and 5-fluorour- G0
acil (5-FU). Figure 4.5
Rationale for the combination of gemcitabine and
Vinca alkaloids and taxanes carboplatin.
Either derived from plant alkaloids or synthetic
derivatives of them, they interfere with mitotic spindle of dividing cells. Taxanes cause mitotic
spindle assembly in M phase causing metaphase arrest by forming abnormal spindle fibres. One
arrest. Vinca alkaloids are extracts of the peri- of the common toxicities of these agents is
winkle plant. They bind to microtubular proteins peripheral neutropathy. Examples are vincristine
which are essential to the formation of the mitotic and paclitaxel.
34
PRINCIPLES OF SYSTEMIC THERAPY 4
Purine synthesis Pyramidine synthesis
Box 4.11: Sanctuary sites
Consultant:
Date PREMEDICATION Dose Route Rate Special Instructions Drs sig Time Check Admin
Date CYTOTOXIC DRUGS Dose Route Rate Volume Special Instructions OP Pharm Batch Number Start Time Admin
Cyto Pharm Date Stop Time Check
Day 1
Figure 4.7
A sample chemotherapy prescription chart.
36
PRINCIPLES OF SYSTEMIC THERAPY 4
Box 4.13: Pre-prescribing chemotherapy checks Box 4.14: Prescribing carboplatin: AUC and
the Calvert formula
• Is the allergy box filled in and does it contain
anything which causes concern for the The required dose is calculated by the following
chemotherapy or supportive drugs? formula:
• Is this the correct chart for that treatment? Dose = AUC × (GFR + 25)
• Is the performance status appropriate for the type where AUC is the area under the curve. It refers to
and intent of treatment? increasing dosages which have an increasing effect
• Are the height and weight accurate and up to until the effect plateaus and any increase in dose
date? results only in increased toxicity. In general AUC
• Is the body surface area correct? (Have your own values are in the range 5–7 in adults but up to 9 in
calculator or check on a computer). children.
• Has the baseline imaging been performed and the
mid treatment imaging booked to allow
assessment of response?
• Has a GFR been performed (for drugs such as
carboplatin, ifosfamide)?
dose for an individual, but for most drugs the use
• Has a MUGA been performed (for anthracyclines
of surface area calculations continue.
and herceptin)? Does the next MUGA need to be Some drugs are not metabolized in the same
booked? way. Carboplatin is the classical example of
• Has the patient previously received a similar drug this type of drug. During the original phase I
which limits the dose and (e.g. anthracyclines) has
the cumulative dose been recorded?
studies the dose limiting toxicity of carboplatin
• Are the appropriate blood results available and was thrombocytopenia. However, unlike other
are they in the correct range for the drugs being drugs, there appeared to be no relationship
given? Has there been a significant deterioration between the dose administered according to mg/
in a blood result which could be attributable to
one of the chemotherapy drugs? m2 and the degree of thrombocytopenia. It was
• Does the dose need modifying on the next cycle known that carboplatin is excreted almost
due to toxicity? (you may only know this after entirely by the kidney and Calvert and colleagues
reviewing the patient). then realized that the drug’s toxicity was related
• Does the patient have significant co-morbidities to the glomerular filtration rate (GFR). This gave
which due to the toxicities of the chemotherapy
may require dose reduction? rise to the Calvert formula which is used to pre-
scribe carboplatin (Box 4.14). It requires knowl-
edge of the GFR, ideally calculated by nuclear
medicine scan.
Some recent drugs, especially the small mole-
especially important in that case. Before the chart cules such as the tyrosine kinase inhibitor, imat-
is written, given that chemotherapy is toxic, a inib, are given as a standard dose which is the
number of calculations and checks have to be same for all patients. For imatinib the standard
made. These are summarized in Box 4.13. dose is 400 mg per day.
Chemotherapy is generally calculated based
on the surface area of an individual. This is an Routes of chemotherapy administration
historic phenomenon based on the extrapolation Systemic treatments can be given by a number of
of doses administered to animals (given as dose routes. Chemotherapy is usually given intrave-
per surface area) in preclinical models. However nously (IV) but some drugs can be given orally
the initial human surface area calculations were and some as both IV and orally. For some intra-
based on hemi-body moulds created from wrap- venous drugs there are several ways to administer
ping brown paper around a small number of it. For example, 5-fluorouracil (5-FU) can be
cadavers some of whom were children. In addi- given as a daily bolus for 5 days every 3 weeks
tion many other drugs are given to adults in a (Mayo regimen) or as a 48 hour continuous
flat (or standard) dosing such as paracetamol. infusion every 2 weeks (modified de Gramont)
Some studies suggest that giving everyone the (p. 165). This drug needs prolonged or repeated
dose calculated for the average surface area of exposure as it acts in S phase but these two
1.7 m2 was as effective as calculating the specific methods produce different toxicity profiles with
37
I BASIC PRINCIPLES OF CANCER MANAGEMENT
the Mayo regimen producing more significant monitored. It also allows the clinician to record
stomatitis and myelosuppression whereas modi- dates of future scans which can be reviewed prior
fied de Gramont produces significant palmo- to further treatment. Toxicity should be scored
planter erythema. Although many clinicians according to an internationally accepted system
believe the two methods to be equally effective, such as the National Cancer Institute Common
randomized trials have not been performed for Toxicity Criteria and Adverse Events version 3
all cancer types so that some oncologists will only (NCI CTCAE v3) which can be found at: http://
prescribe the method used in the original trials. www.fda.gov/cder/cancer/toxicityframe.htm.
Some drugs can also given either intrathecally
Dose adjustment
(Box 4.15 and Figure 4.8) or via the intraperi-
toneal route (Box 4.16 and Figure 4.9). Although supportive drugs to minimize side
More recent drugs again have a range of effects are usually given prophylactically signifi-
administration routes from intravenous, e.g. cant toxicity can occur. Initially the aim would
monoclonal antibodies, to subcutaneous, e.g. be to change or add to the supportive drugs to
vaccines, to oral, e.g. tyrosine kinase inhibitors. reduce toxicity, e.g. different or additional anti-
Patients often prefer an oral route of administra- emetics for nausea control, or use of G-CSF to
tion or a short intravenous infusion to minimize prevent neutropenic sepsis. If the toxicity has
their time in the hospital but they need to under-
stand that toxicities from oral drugs (e.g. capecit-
abine) can be as great as those from intravenous Ommaya reservoir tap
drugs and that oral chemotherapy is not a ‘gentle
option’. Scalp Syringe
Toxicity recording Brain
Needle
In order to minimize side effects and to evaluate Reservoir tap
how changes in treatment have affected these
side effects or the patient’s symptoms due to the
cancer, accurate recordings of drug toxicity (Box
Cerebrospinal
4.17) must be made. Ideally this should be fluid
recorded on a flow chart so that progress can be
38
PRINCIPLES OF SYSTEMIC THERAPY 4
Toxicity chart
Trial name:
Treatment protocol: Date: Cycle:
Toxicity: 0 1 2 3 4 0 1 2 3 4
Performance status Neuropathy
Fatigue Stomatitis (oral)
Nausea/vomiting Alopecia
Diarrhoea Cardiac
Constipation Bilirubin
Haematological (max) ALT
Figure 4.10
Example of a toxicity chart.
Complementary therapy interactions cific to the cancer, whereas others have broader
Many patients, especially with advanced cancer, actions.
take additional treatments or complementary
therapies. Each hospital has a different policy Hormone treatments (Table 4.1)
about whether patients are allowed to take com- The observation that breast cancer can regress
plementary medicines alongside their chemother- after oophorectomy and that prostate cancer can
apy. However it is important to take a full history be controlled by orchidectomy suggested that
of these and ensure that certain complementary some cancers may be hormone sensitive. Since
treatments which can interact with chemother- the observation that surgical measures to reduce
apy or other drugs. A specific example is St hormone levels were effective, drugs were devel-
John’s wort which many patients take for mild oped which would achieve a similar effect. In the
depression. It is metabolized by the cytochrome 1990s dramatic improvements in breast cancer
p450 isoenzymes, especially CYP 3A4 and can survival were seen with the introduction of the
significantly interfere with the doses of several anti-oestrogen, tamoxifen, to women with oes-
systemic treatments including etoposide and the trogen receptor positive breast cancer. This led
tyrosine kinase inhibitor, imatinib. to the developments of other drugs which target
the production of oestrogen (such as the aro-
Other classes of matase inhibitors) as well as examining the role
of hormone treatments in other hormone sensi-
systemic therapy tive cancers such as prostate cancer. These are
Although chemotherapy has been the mainstay discussed in more detail in the breast and pros-
of systemic therapy, the development of alterna- tate chapters (Chapters 10 and 12). In general
tive methods of treatment has occurred in the last these treatments are taken for many years to
20 years, many of which have made significant prevent recurrence or control the disease and
improvements in survival. Some of these have can be effective without chemotherapy. In fact
been developed against a particular target spe- there is evidence to suggest that they should not
40
PRINCIPLES OF SYSTEMIC THERAPY 4
be given alongside chemotherapy, particularly Other antibodies have been developed with a
for anti-oestrogen therapies in breast cancer as wider range of action such as bevacizumab which
it is thought that these cytostatic agents (e.g. targets the vascular endothelial growth factor
tamoxifen) causes cell cycle arrest preventing the receptor. This has shown some improvement in
action of chemotherapy which is dependent on survival in several cancers, which are covered in
rapidly dividing cells for its action. appropriate chapters. The toxicities of these
drugs are different to cytotoxics with allergic
Bisphosphonates reaction being one of the most significant.
Bisphosphonates represent a good example of an
adjunct in the treatment of cancer. These agents Immune therapy
were originally developed to treat osteoporosis In addition to antibodies, which may be consid-
but have been shown to be active in stabilizing ered an immune treatment, there are also a group
bony metastases in a number of malignancies and of drugs which act by modifying the immune
have also been shown to reduce visceral metas- response of the host (patient) in general, rather
tases in breast cancer. The exact mechanism for than targeting a specific molecule or receptor.
this latter activity is unknown. They can be given Examples in this group include interferons
intravenously or orally. Examples include pamid- and interleukins. High dose interferon has been
ronate and zoledronic acid. used in melanoma (p. 235) and interleukin-2 has
some efficacy in renal cell carcinoma (p. 176).
Antibodies Interferon and interleukin both produce systemic
Antibodies are immunoglobulins produced by side effects of an immune response which resem-
plasma B cells in response to antigens. Several ble a viral infection, e.g. fever, fatigue, myalgia
tumour cells express specific antigens which are and headache.
not produced by normal cells and these have been
the target for the development of monoclonal Small molecules
antibodies to these specific antigens. To date the Small molecules are drugs developed with a
most successful drugs in this class are rituximab specific target, usually to a cellular pathway or
and transtuzumab. Rituximab is directed against molecule within that pathway. Examples include
CD20 found on B cells and the addition of tyrosine kinase inhibitors (TKIs) and mTOR
rituximab to standard chemotherapy for B cell inhibitors. TKIs can be specific to a particular
lymphomas has significantly improved survival. tyrosine kinase, e.g. imatinib, or may have activ-
Transtuzumab has been developed to target the ity across a number of kinases, e.g. sunitinib.
HER-2 receptor which is found in 25% of breast Figure 4.11 shows an example of a tyrosine kinase
cancers. Initially used in metastatic disease but signalling pathway using c-KIT. Once the recep-
then in the adjuvant setting for women with tor is bound it sets off a cascade leading to further
HER-2 receptor positive breast cancer, this has cell proliferation. Some of the cascade steps are
also seen a significant improvement in survival. common to pathways involved in many cancers
41
I BASIC PRINCIPLES OF CANCER MANAGEMENT
such that effective inhibitors to one protein could However for some tumours or in the adjuvant
be useful drugs in a range of cancers. setting RECIST terms may not adequately
describe response. In adjuvant treatment where
Systemic radiotherapy the aim is to cure, the overall survival and some-
In general radiotherapy is directed against a spe- times the progression free survival are the key
cific site in the body. Generally a radioisotope indicators of success. For clinical trials this may
scan is done to assess the uptake by the specific take many years to assess so sometimes other
tumour prior to giving definitive radioablative indicators are used. These include pathological
dose (for example, see p. 282). Doses can be response for tumours such as bone tumours or
repeated several times but there is a risk of late breast cancer in whom the patients have been
myelodysplastic syndromes. given neoadjuvant treatment. The degree of cell
necrosis or pathological response reflects progno-
Assessment of response sis in these groups. For gastrointestinal stromal
tumours which have been treated with a targeted
to treatment treatment, imatinib, the response assessed by
Systemic treatments are often given to treat meta- RECIST criteria may not reflect the true response.
static disease or neoadjuvantly prior to surgery. For these tumours, and other sarcomas there may
In these cases an assessment needs to be made not be an obvious reduction in the size of the
to ensure that the treatment is effective (Figure tumour, and yet the patient feels better. Radio-
4.12) so that an alternative treatment may be logically this is usually accompanied by a change
considered and to avoid inappropriate toxicity. in the density of the tumour becoming more
Most often this assessment is made radiologically cystic and less dense. For gastrointestinal stromal
(e.g. by CT or MRI) by Response Evaluation tumours new criteria (Choi criteria) have been
Criteria in Solid Tumours (RECIST) criteria applied to these tumours to take these changes
(Boxes 4.18 and 4.19, Table 4.2). An under- into account (p. 263).
standing of these terms is necessary in the man- In addition, particularly in the context of clini-
agement of all cancers, even if alternative means cal trials the term Clinical Benefit is used. This
of assessment may also be used (e.g. see section takes into account not only the obvious CR and
on gastrointestinal stromal tumours, p. 261). PR RECIST responses but also those tumours
Figure 4.11
Receptor C–KIT
Simplified diagram to show signalling
pathway of the tyrosine kinase c-KIT
and the cascade that follows once
Cell membrane the receptor is bound. Targets are
being developed to several proteins
Cytoplasm along this pathway including raf,
Tyrosine kinase 1 SHP2 MEK, P13K and AKT.
Raf
AKT STAT
42
PRINCIPLES OF SYSTEMIC THERAPY 4
A B
Figure 4.12
CT scan showing a large recurrent uterine sarcoma prior to chemotherapy (A) and after 6 cycles of treatment
demonstrating a complete response (B). The patient went on to have radical radiotherapy and remains free of disease
6 years later.
Box 4.18: Definition of lesions for RECIST Box 4.19: Confirmation of response by
evaluation RECIST criteria
• Measurable lesions – lesions that can be accurately • The main goal of confirmation of objective
measured in at least one dimension with longest response is to avoid overestimating the response
diameter ≥20 mm using conventional techniques rate observed. In cases where confirmation of
or ≥10 mm with spiral CT scan. response is not feasible, it should be made clear
• Non-measurable lesions – all other lesions, when reporting the outcome of such studies that
including small lesions (longest diameter <20 mm the responses are not confirmed.
with conventional techniques or <10 mm with • To be assigned a status of PR or CR, changes in
spiral CT scan), e.g. bone lesions, leptomeningeal tumour measurements must be confirmed by
disease, ascites, pleural/pericardial effusion. repeat assessments that should be performed no
less than 4 weeks after the criteria for response
are first met. Longer intervals as determined by
the study protocol may also be appropriate.
The future of systemic • Finding the best way to assess response for new
therapies.
treatments (Box 4.20) • Learning the best way to combine new therapies
either with other new agents or with
This chapter has highlighted the growth and chemotherapy.
growing diversity of systemic treatments for
cancer from early fortuitous discoveries of chem-
otherapeutic agents to the idea of targeted treat- combine drugs, e.g. cytotoxic with a small mol-
ments. The future of systemic therapies lies in the ecule or a number of targeted small molecules
development of more specific and less toxic drugs, without chemotherapy. This, along with the
and the methods to best assess them. It also changing arena of clinical trials, represents one of
remains to be discovered how is the best way to the great challenges for the future oncologist.
43
I BASIC PRINCIPLES OF CANCER MANAGEMENT
44
Cancer genetics
E Copson
5
Introduction Inheritance of two alleles each carrying a
mutation within the same tumour suppression
The last two decades have seen rapid expansion gene is extremely rare and can result in a differ-
in our understanding of the genetic origins of ent and more severe phenotype than monoallelic
cancers. Between 5–10% of cancers are due to mutation inheritance. For example, inheritance
inheritance of alterations in genes that confer a of one BRCA2 mutation from a single parent is
high life-time risk of certain malignancies, such associated with breast/ovarian cancer whilst bial-
as breast, colorectal cancer (see Table 5.1), and lelic BRCA2 mutations are a cause of Fanconi’s
very rare tumours. anaemia.
Most of these so-called ‘hereditary cancer pre-
disposition syndromes’ are due to mutations
within a single allele (copy) of a tumour suppres- Diagnosis
sor gene inherited in an autosomal dominant A hereditary predisposition towards developing
fashion. malignancy is characterized by young age at
presentation, an increased incidence of bilateral
Mechanisms of inherited cancer tumours, more than one primary tumour site and
Tumorigenesis involves contributions from two a family history of specific cancers. The first step
classes of genes. Oncogenes control cell growth requires a detailed family history containing all
and proliferation under normal circumstances, maternal and paternal cases of malignant disease,
but once inappropriately activated or overex- with particular emphasis on age of onset and the
pressed can promote rapid clonal expansion. site of the primary tumour. Wherever possible,
Tumour suppressor genes normally inhibit verification of these diagnoses should be gained
abnormal cell proliferation; reduced expression from pathology records.
of these genes can result in uncontrolled cell Analysis of DNA for mutations in known
division. Almost all hereditary cancer predisposi- cancer predisposition genes is performed in a
tion syndromes are due to the inheritance of a limited number of specialist molecular genetics
germline mutation in a tumour suppressor gene. laboratories using DNA derived from a periph-
The inherited mutation inactivates one copy of eral blood sample. The gold standard is direct
the gene and the subsequent development of a sequencing of the whole gene but this is generally
mutation in the remaining ‘normal’ copy of the not commercially viable for very large genes such
gene results in failure of a cell to produce the as BRCA1/2. Screening for pathogenic mutations
tumour suppressor protein (Knudson ‘two-hit’ is therefore often limited to analysis of coding
hypothesis – see Figure 5.1). regions only, or regions of the genes which carry
The concept of heritable cancer predisposition the greatest density of reported mutations. Addi-
was originally developed from observations of an tional tests are required to detect gene deletions.
earlier age of onset of retinoblastoma in patients A full screen for unknown mutations will take
with a positive family history. several weeks to process. ‘Predictive testing’, in
Table 5.1: The risk of common cancers associated with inherited cancer syndromes
Genes associated with
Tumour site relative risk of >5.0 Cancer syndrome
Breast BRCA1/2 Hereditary breast/ovarian cancer syndrome
TP53 Li–Fraumeni syndrome
PTEN Cowden syndrome
CDH1 Hereditary diffuse gastric cancer syndrome
STK11 Peutz–Jeghers syndrome
Colorectal APC Familial adenomatous polyposis
MUTYH
MLH1 Lynch syndrome (hereditary non-polyposis colon cancer)
MSH2
MSH6
PMS2
Pancreatic BRCA2 Hereditary breast/ovarian cancer syndrome
BRCA1
MSH2 Lynch syndrome (hereditary non-polyposis colon cancer)
MLH1
STK11 Peutz–Jeghers syndrome
TP53 Li–Fraumeni syndrome
PRSS1 Hereditary pancreatitis
SPINK1 Familial atypical mole malignant melanoma syndrome (FAMMM)
CDKN2A
Prostate BRCA2 Hereditary breast/ovarian cancer syndrome
Endometrial MLH1 Hereditary non-polyposis colon cancer
MSH2
MSH6
PMS2
Ovarian BRCA1/2 Hereditary breast/ovarian cancer syndrome
MLH1 Lynch syndrome
MSH2
which genetic testing is directed at establishing cancer predisposition syndrome receives expert
whether or not an individual has a specific muta- counselling prior to testing and on receiving
tion carried by another family member, is faster. their result. Patients must be advised that failure
to detect a mutation does not always mean
that no mutation is present and that further results
Genetic counselling may become available in the future when genetic
It is essential that any individual considering testing may become more sophisticated. The roles
undergoing genetic screening for an inherited of genetic counselling are summarized in Box 5.1.
46
CANCER GENETICS 5
One parent carrying Figure 5.1
mutation in tumour Comparison of tumourigenesis
suppressor gene pathways in carriers and non-
carriers of a mutation in a tumour
Tumourigenesis suppressor gene – the Knudson
Non-carrier “sporadic” ‘two hit theory’.
Box 5.1: The roles of the genetic counsellor Box 5.2: Women at increased risk of breast
cancer
• Explaining the difference between sporadic and
inherited cancer risk. Women are generally considered to be at increased
• Facilitating the documentation of a detailed risk of breast cancer if they have:
family history. • One close relative diagnosed with breast cancer
• Explaining alternative options to genetic testing, under the age of 40, or
i.e. approximation of personal risk on basis of • Two close relatives (in same blood line) diagnosed
family history. under the age of 50, or
• Explaining the risk of passing on a mutation to • Three or more close relatives diagnosed at any
offspring. age
• Identifying the most appropriate family member
to undergo genetic testing.
• Ensuring individuals understand the implications
of a positive finding of a mutation in a cancer condition (Box 5.2). Approximately 5% of all
predisposition gene result, including current
recommendations for screening and prophylactic breast cancer cases are thought to be due to
interventions. highly penetrant autosomal dominant cancer
• Ensuring individuals understand the implications predisposition syndromes (Figure 5.2). The two
of a negative result, including population most frequent highly penetrant breast cancer sus-
screening for common cancers.
ceptibility genes are BRCA1 and BRCA2 where
• Ensuring individuals understand the implications
of an ambiguous result. in some families lifetime penetrance for breast
• Helping to communicate test results to other cancer can reach 80% but penetrance is clearly
members of the family. modified by other genetic and environmental
• Ensuring individuals understand that genetic factors. Much rarer syndromes including Li–
testing may result in discrimination by insurance Fraumeni, Cowden, Peutz–Jeghers and heredi-
companies.
tary diffuse gastric cancer (HDGC) syndromes
• Focusing on family health.
can be recognized either by other clinical mani-
festations or by the pattern of cancers in the
family (Table 5.2). Lifetime breast cancer risk for
The remainder of this chapter will look in
carriers of these mutations is in the order of
more detail at inherited breast and colorectal
20–100%. It is likely that most familial clusters
cancer, and the most common cancer predisposi-
of breast cancer are the consequence of multiple
tion syndromes.
low penetrance cancer susceptibility genes acting
in conjunction with environmental factors.
Inherited breast cancer Within the last few years a number of large case-
Studies indicate that 20–30% of breast cancer control studies have identified variants in DNA
patients report a positive family history of this repair genes (e.g. CHEK2, ATM, BRIP1 and
47
I BASIC PRINCIPLES OF CANCER MANAGEMENT
Figure 5.2
No family history of breast The importance of inherited factors in
cancer (sporadic) the aetiology of breast cancer.
Table 5.2: Cancer predisposition syndromes associated with an increased risk of breast cancer
Clinical syndrome Gene Population frequency Breast cancer risk Other component tumours
Hereditary breast BRCA1 1 : 1000 40–80% Ovarian
cancer lifetime risk
Hereditary breast BRCA2 1 : 750 40–80% Ovarian
cancer lifetime risk Prostate
Pancreatic
Li–Fraumeni TP53 <400 families 30–60% Sarcoma
worldwide by age 45 Brain tumours
Adrenocortical tumours
Leukaemia
Cowden PTEN 1: 300,000 25–50% Thyroid
lifetime risk Endometrial
PALB2), which double the risk of breast cancer. Genetics and pathology
The population prevalence of these variants BRCA1 and BRCA2 act as classical tumour sup-
seems to be in the order of 1–5%. pressor genes; inheritance of mutations occurs in
Hereditary breast/ovarian cancer an autosomal dominant fashion and loss of the
unmutated allele is found in tumour specimens.
syndrome
The precise functions of the BRCA1 and BRCA2
Clinical features gene products remain unclear but evidence indi-
See Table 5.3. cates that these proteins are involved in DNA
48
CANCER GENETICS 5
Table 5.3: Clinical features of BRCA1 and BRCA2 hereditary breast/ovarian cancer syndrome
Feature BRCA1 BRCA2
Gene locus Chromosome 17 Chromosome 13
Population frequency 1 in 400 1 in 400
Increased to 1 in 50 in Ashkenazi Jews Increased to 1 in 50 in Ashkenazi
Jews
Inheritance Autosomal dominant Autosomal dominant
Lifetime risk of breast cancer 40–80% 40–80%
(females)
Pathological features of breast Typically high grade with lymphocytic No typical phenotype
tumours infiltrate and pushing tumour margins,
ER, PR, HER-2 negative
Lifetime risk of ovarian cancer 40–60% 10–20%
Other associated tumours Pancreatic Male breast cancer
Pancreatic
Prostate
repair and transcription regulation, with addi- Box 5.3: Clinical features indicative of a possible
tional roles in cell cycle checkpoint control and underlying BRCA1/2 mutation in breast cancer
cytokinesis. Sporadic breast cancers do not patients
contain BRCA1 mutations and very rarely dem- • Young age at diagnosis (<35 years)
onstrate BRCA2. • Strong family history of breast cancer, e.g.
• 2 first degree relatives with breast cancer, with
Diagnosis average age at diagnosis <50 years
A number of scoring systems exist to estimate the • ≥3 first or second degree relatives with breast
cancer, diagnosed an average age of <60
likelihood of a family carrying BRCA1 or BRCA2
• Family history of breast and ovarian cancer
mutations, e.g. the Gail model. Most cancer
• Family history of male breast cancer
genetic units recommend screening for BRCA 1/2
• Askenazi Jewish ethnicity
mutations if the chance of carrying a mutation is
• Bilateral breast cancer (if first diagnosed at <50
estimated at more than 20%. Oncologists should years)
consider the possibility of an underlying BRCA1/2 • (High grade ER/PR/HER2 negative tumour)*
mutation in breast cancer patients with the clini-
cal features listed in Box 5.3. *BRCA1 only.
Genetic counselling
Predictive genetic testing for Testing for BRCA1/2 Declines testing for
known BRCA1/2 mutation mutation BRCA1/2 mutation
Enter breast screening Further Consider testing for Offer breast cancer
as per general population genetic counselling other hereditary screening as appropriate
cancer syndromes for family history
Standard management
of breast cancer /
clinical trials
** See text
Figure 5.3
Algorithm for management of potential BRCA1/2 mutation carrier.
been recommended for all patients with known Treatment of BRCA associated breast cancer
BRCA1 or BRCA2 mutations for some years. The management of potential BRCA1/2 muta-
The 2006 National Institute for Health and Clin- tion carriers is summarised in Figure 5.3 and
ical Excellence (NICE) guidelines for the man- discussed in Chapter 10 on breast cancer.
agement of familial breast cancer include a
recommendation for annual MRI surveillance of
all BRCA1/2 mutation carriers aged 30–49 as
Inherited colorectal cancer
this method of screening has proved more sensi- Approximately 5% of cases of colorectal cancer
tive then mammography. Screening for ovarian are due to an underlying hereditary cancer syn-
cancer remains under investigation. drome, with an increased representation amongst
50
CANCER GENETICS 5
* See Table 5.4 for HNPCC related cancers. Familial adenomatous polyposis
Clinical features
FAP is characterized by the development of
hundreds of colonic adenomatous polyps by the
51
I BASIC PRINCIPLES OF CANCER MANAGEMENT
52
CANCER GENETICS 5
Cowden syndrome
Cowden syndrome is an autosomally dominant
Table 5.6: Component tumours of classic inherited cancer predisposition syndrome charac-
Li–Fraumeni syndrome and other tumours terized by multiple hamartomas and a high risk
associated with LFS
of thyroid and breast cancers, with other tumours
Classic Li–Fraumeni Other tumours
component tumours associated with LFS also at increased frequency.
A clinical diagnosis of Cowden syndrome is
Breast carcinomas Leukaemia
Soft tissue and bone Lung currently made according to the International
sarcomas Gastrointestinal Cowden Consortium operational criteria (see
Brain tumours Ovary Table 5.7).
Childhood adrenal cortical Lymphomas
carcinoma Paediatric tumours Genetics
(Choroid plexus tumours) Melanoma
Prostate
In 80% of Cowden syndrome families, the under-
Pancreatic lying mutation is in the PTEN (protein tyrosine
(+ many others) phosphatase with homology to tensin) gene,
located on chromosome 10. PTEN encodes
a phosphatase which mediates cell growth,
54
CANCER GENETICS 5
proliferation, cell cycle arrest and/ or apoptosis, tions. Pre-implantation genetic testing has been
hence acting as a tumour suppressor. performed for this condition.
Incidence Management
The incidence of Cowden syndrome is estimated Early identification of a RB1 mutation in the
to be 1 in 200,000. Inheritance of a germline child of an affected parent can permit early diag-
PTEN mutation is associated with a 25–50% nosis of retinoblastoma (with the aim of salvage
lifetime risk of breast cancer in females. of the eye), by regular fundoscopic examinations
from birth. An increased frequency of second
Clinical management tumours has been associated with irradiation.
There is currently no consensus on screening.
Individuals with known or suspected PTEN muta- Von Hippel–Lindau syndrome
tions should be referred to their local cancer
Von Hippel–Lindau (VHL) is a rare hereditary
genetics unit to seek up to date advice about
cancer syndrome consisting of haemangioblasto-
surveillance.
mas of the retina and central nervous system
(particularly cerebellar, medullary and spinal
Retinoblastoma sites), associated with phaeochromocytomas
Retinoblastoma is a rare primary malignant and clear cell renal carcinomas. It is inherited
tumour of the retina. Unilateral tumours usually in an autosomal dominant fashion. Hereditary
present before the age of 4 years (median age 18 phaeochromocytomas present earlier and are
months) whilst bilateral tumours present even more frequently multiple than sporadic phaeo-
earlier (median age 13 months). Only 5–10% of chromocytomas but are less likely to undergo
patients have a positive family history. Second malignant transformation. Renal tumours occur
primary tumours are seen in a quarter of surviv- in 25% of VHL families, presenting at a median
ing patients with hereditary retinoblastoma, age of 45 years. They are also more likely to be
occurring up to 40 years after the initial event. bilateral than sporadic renal carcinomas.
These include sarcomas, melanoma, brain
tumours, breast cancers and leukaemia. Genetics
The VHL gene is sited on chromosome 3. Muta-
Genetics tions or deletions of the VHL gene are found in
Germline mutations of the RB1 gene on chromo- virtually all clinically diagnosed VHL families
some 13 are found in over 80% of hereditary or using modern genetic diagnostic methods.
bilateral cases of retinoblastoma and are inher-
ited in an autosomal dominant fashion. The RB1
Incidence
gene encodes a protein which acts as a transcrip- VHL syndrome is estimated to affect 1 in 35,000
tion regulator, suppressing the expression of individuals.
genes required for cells to progress through the
Clinical management
cell cycle and into mitosis. Penetrance of RB1
mutations is variable and ‘skipping’ of genera- Screening for VHL tumours should be as set out
tions may be observed. in Table 5.8. Pre-natal and pre-implantation
genetic diagnosis is now available for VHL when
Incidence a mutation has been identified in a family.
Retinoblastoma is very rare, with an incidence of
1 in 20,000 births worldwide. Approximately Multiple neuroendocrine
45% of cases are hereditary.
neoplasia syndromes
Diagnosis The multiple endocrine neoplasia (MEN) syn-
Identification of mutations within the retinoblas- dromes are autosomal dominant disorders char-
toma gene is complicated by the large size of this acterized by the development of multiple benign
gene, with numerous different reported muta- and malignant tumours of endocrine glands. The
55
I BASIC PRINCIPLES OF CANCER MANAGEMENT
key clinical features of MEN1, MEN2A and oncogene RET which codes for a transmembrane
MEN2B are summarized in Table 5.9. receptor tyrosine kinase. The position of the
mutation within the RET gene seems to deter-
Genetics mine whether the clinical features of MEN2A or
MEN1 is caused by mutations in the menin gene MEN2 develop.
located on chromosome 11. Menin is a tumour
suppressor gene which interacts with a number of
nuclear proteins including transcription factors. Incidence
Both MEN2A and MEN2B are associated The incidence of MEN is between 0.2–2 per
with mutations of the chromosome 10 proto- 100,000.
56
CANCER GENETICS 5
57
Late effects of cancer
treatment and survivorship
HM Hatcher
6
Introduction Medical late effects
In the UK the estimated numbers of people living Late effects are dependent upon the original
after a cancer diagnosis range from 1–1.5 million. cancer, its treatment, the family genetics and
With increasing cancer incidence and survival it the developmental stage of the individual when
is thought that this will rise to represent 1.5– treated for cancer. Box 6.1 lists some of the
2.5% of the adult population. The 5-year sur- major effects that can occur.
vival for children diagnosed with cancer is now
79% and for adult cancers is 64%. There are a Second malignancy
number of medical, psychological, social and Second malignancies are the second most
economic issues which can significantly contrib- common cause of death (after the primary cancer)
ute to a patient’s morbidity and mortality after in those diagnosed with cancer, and can either be
a diagnosis of cancer. This chapter aims to over- a solid tumour or haematological (leukaemia or
view these issues. myelodysplastic syndromes). The risk of second
malignancy is increased with combinations of
Paediatric oncology and chemotherapy and radiotherapy but is also
dependent upon the underlying genetics of the
the history of survivorship individual and the sensitivity of the tissue irradi-
The median age of diagnosis of cancer in the ated (Table 6.1).
general population is 70 years but the most sig- In general secondary solid tumours arise in
nificant increase in survival rates in the past 30 sites of previous radiotherapy, especially if chem-
years has been in the area of paediatric oncology. otherapy was also given. The total dose of radio-
Many children were treated in a clinical trial therapy delivered as well as the type and energy
such that the initial survivorship data was gained of the treatment and treated volume determine
from paediatric patients. The medical sequelae the risk. The tissues most likely to give rise to a
of cancer treatment as a child have profound secondary malignancy following radiotherapy
consequences due to the current average life are bone marrow, thyroid, breast and soft tissues
expectancy. (sarcomas) (Box 6.2).
Given the dependence of children upon their Leukaemias and myelodysplastic syndromes
parents and the developmental needs of a growing have been found to relate to previous chemo-
child some of their psychosocial survivorship therapy treatment, especially with etoposide,
issues are different to those of adults. In addition anthracyclines and alkylating agents. These leu-
the families of children treated for cancer are kaemias are characterized by a chromosomal
significantly affected and many parents or sib- abnormality at 11q23, tend to occur within 2
lings of childhood survivors suffer long-lasting years of primary treatment and have a poor prog-
psychological and social consequences even when nosis. Intensive regimens for Ewing’s sarcoma or
the clinical outcome has been good. rhabdomyosarcoma have a risk of up to 20% at
58 © 2011, Elsevier Ltd
DOI: 10.1016/B978-0-7234-3458-0.00011-7
LATE EFFECTS OF CANCER TREATMENT AND SURVIVORSHIP 6
Box 6.1: Medical late effects of treatment Box 6.2: Learning points
• Secondary tumours are often very aggressive and
Second malignancy e.g. leukaemia, sarcoma
do not respond as well to treatment as primary
Chronic health e.g. breathlessness, fatigue
tumours of the same histology.
conditions
Cardiological e.g. arrhythmias • In Ewing’s sarcoma, radiotherapy with greater
Neurological e.g. peripheral neuropathy than 54 Gy is associated with second malignancy
Pulmonary e.g. fibrosis but treatment with less than 45 Gy is associated
Endocrine e.g. GH deficiency with local recurrence.
Fertility e.g. premature ovarian failure
Bone e.g. osteoporosis
Renal e.g. hypomagnesaemia
Figure 6.1
Coronal T2 weighted MRI through the pelvis showing
abnormal bony texture in the left acetabulum with an
irregular margin (arrow) in contrast to the normal right
side which shows normal marrow architecture. The left
sided mass is an osteosarcoma.
Box 6.4: Fertility options for women Box 6.5: Timing of pregnancy after
cancer treatment
Currently options are limited for women who require
intensive treatment or pelvic radiotherapy and many Timing of pregnancies for women after treatment for
are costly and not provided by the NHS. Examples cancer is problematic. If the patient remains fertile
include: they will be advised to start their families earlier
• In some cases ovarian suppression with LHRH rather than later. However, as the risk of relapse
(leuteinizing hormone releasing hormone) during occurs in the first few years after treatment for most
treatment, e.g. alongside hormonal treatment for cancers, and the general advice is to wait at least 2
their breast cancer (e.g. tamoxifen) rather than years after completion of treatment. However a large
removal of the ovaries, allows preservation of study showed no increase in rates of relapse for
fertility for some women. breast cancer patients who became pregnant within
a few years of diagnosis.
• Frozen embryos can be successful but the woman
needs to be well enough to defer treatment for a Patients still on maintenance therapy such as
few weeks to complete an IVF cycle and egg tamoxifen should be advised that pregnancy is not
retrieval. Problems associated with this include the recommended whilst taking these drugs due to the
need for a long-term partner to donate sperm to risk for the developing baby.
create the embryos, which is often not the case
for younger patients. An additional problem is
that if the father later withdraws consent for
the embryos to be implanted they must be
destroyed. Box 6.6: Risk factors for hormone induced bone
loss and fracture
• Oocyte removal after an IVF cycle has also been
used, without immediately creating an embryo. • Treatment with hormone manipulation, e.g.
Frozen eggs have now given rise to a very few aromatase inhibitor or anti-androgen
pregnancies and the process is still experimental • Smoking
but the technique is improving so should be
discussed if available. • Heavy alcohol use
• Research is ongoing using extracted frozen • Family history
immature eggs which do not require the time • Bone metastases
delay of a fertility treatment cycle but have yet to • Increasing age
be proven to be a reliable option.
• Still in the experimental stages is the storage of
ovarian tissue which can later be reimplanted into
the patient after treatment.
chemotherapy such that calcium supplementa-
• Surrogacy can be considered if the woman
remains fertile but the uterus is unable to carry a
tion or bisphosphonates are frequently required
full-term pregnancy due to surgery or to prevent fractures. To minimize the risk of
radiotherapy. fractures from significant osteoporosis patients
• Egg donation may be a possibility with or without should be screened for osteoporosis prior to com-
surrogacy.
mencing hormonal treatment using a risk assess-
• Adoption can always be considered.
ment and bone mineral density scan. Patients
should be reassessed after 6–12 months on hor-
monal therapy. Those at significant risk should
the offspring, including after bone marrow trans- be advised about calcium in their diet and/or
plantation (Box 6.5). prescribed calcium supplements. At very high
risk or after a fracture, bisphosphonates (orally
Bone late effects or intravenously) should be prescribed.
Bone growth is affected by steroids, chemother- Osteoradionecrosis can occur after high-dose
apy or radiotherapy (Box 6.6). In addition hor- radiotherapy particularly to the head and neck
monal manipulation with aromatase inhibitors region. Pre-treatment dental assessment and cor-
or anti androgen therapy can have profound con- rective procedures are important in preventing
sequences on bone loss. Osteopenia occurs fre- this side effect.
quently at the end of treatment with combination Osteonecrosis is a known complication of
chemotherapy. In ER positive breast cancer treatment for leukaemia, lymphoma or bone
where patients may have chemotherapy followed marrow transplantation and is thought to be due
by an aromatase inhibitor, the hormonal treat- to high-dose steroids. It tends to occur most in
ment compounds the osteoporotic effect of the weight bearing bones e.g. femur such that arthro-
61
I BASIC PRINCIPLES OF CANCER MANAGEMENT
plasty of the hip joint may be carried out in up neuropathies are most commonly associated with
to 20% patients with femoral head osteonecro- these drugs, especially vincristine. For cisplatin,
sis. Osteonecrosis of the jaw is a recognized com- the symptoms and signs of peripheral neuropathy
plication of treatment with zolendronic acid. can progress even after treatment has finished
Patients should see a dental surgeon prior to which highlights the importance of detecting
starting treatment and zolendronic acid should deterioration.
be stopped and further dental assessments made
if there is any suspicion of dental abscess.
Example box 6.2:
Neurological
A 17-year-old man with medulloblastoma (Figure
Neurological damage can occur either centrally 6.3A) was treated successfully with combined
to the brain itself or to the peripheral or chemo-radiotherapy followed by further
autonomic nerves depending on the tumour and chemotherapy. Although one of his presenting signs
was of unsteadiness this deteriorated significantly
treatment given. For those with CNS tumours, part way through his chemotherapy such that he was
cranial radiotherapy may be given in combina- unable to stand or walk unaided. Recurrence was
tion with chemotherapy which can produce suspected but the MRI (Figure 6.3B) showed no signs
of this but showed scarring consistent with previous
combined peripheral and central neurological radiotherapy. On examination of the nervous system,
deficits. there had been no change in the central features.
Certain chemotherapeutic agents are known However, a significant peripheral neuropathy had
developed with loss of proprioception below the
to be neurotoxic such as platinums and vinca ankles and wrists. Unfortunately, although his MRI
alkaloids. The risk is greater with increasing age, shows no evidence of recurrence 5 years later he has
particularly over the age of 50 and unless specifi- been left unable to walk out of the house unaided
and is unable to work due to his peripheral
cally assessed during treatment, the damage may neuropathy (Box 6.7).
be permanent and debilitating. Painful peripheral
A B
Figure 6.3
MRI FLAIR sequence showing medulloblastoma in right cerebellum prior to radiotherapy (A) and MRI of brain after
radiotherapy when the patient presented with worsening neurological symptoms (B).
62
LATE EFFECTS OF CANCER TREATMENT AND SURVIVORSHIP 6
who have received high-dose steroids, particu- cranial prophylaxis with radiotherapy and the
larly during a period of infection. high doses of corticosteroids used.
Endocrine input is essential in the assessment
of late effects in the follow-up clinic. Chronic health conditions
In addition to specific toxicities the overall health
Obesity of an individual is likely to be affected after treat-
Obesity is a significant problem for survivors of ment for cancer. Chronic health conditions can
cancer, especially those treated in childhood. The include overall health, or any of the above
risks are greatest for those treated with cranial medical conditions which have a chronic effect
radiotherapy, females and those aged 0–4 years on an individual’s health. It also includes symp-
at diagnosis. Survivors of ALL have the greatest toms of chronic fatigue which can cause limita-
risk, thought to be due to a combination of tions of everyday life and persist for many years
after treatment is complete. In a study of child-
hood survivors, chronic health conditions
occurred in almost all patients with 27.5% expe-
Table 6.2: Major cardiac problems associated riencing severe or life-threatening health effects.
with cancer treatments Multiple chronic conditions were also more fre-
Nature of cardiac quent with 37.6% experiencing at least 2 and
problem Causative agent(s) 23.8% experiencing at least three significant
Arrhythmias (many Radiotherapy, multiple co-morbidities.
asymptomatic with chemotherapy agents
impact unknown) e.g. anthracyclines,
taxanes
Causes of death after cancer
Cardiac failure Radiotherapy
treatment
Anthracyclines The main cause of death following a diagnosis of
Trastuzumab
cancer is the cancer itself even after surviving
Coronary artery disease Supra diaphragmatic over 5 years from diagnosis. After recurrence the
and myocardial radiotherapy greatest risk of death is from a second malig-
infarction Anthracyclines
Vincristine
nancy followed by pulmonary then cardiac prob-
lems. In one study the standardized mortality
Note:
hypercholesteralaemia
ratio was 19.4 for second malignancy, 8.2 for
is common after cardiac death and 9.2 for pulmonary death with
chemotherapy other causes at 3.3. The risk of death was great-
est for women, those diagnosed before age 5 and
those with an initial diagnosis of leukaemia or
Table 6.3: Overview of most common endocrine abnormalities and their cause
Endocrine organ
affected Hormones affected Causative agent
Pituitary Growth hormone Radiotherapy to brain (pituitary region)
TSH
FSH/LH
Thyroid Thyroxine Radiotherapy to neck or upper chest >15 Gy (scatter effect)
Gonads Testosterone Radiotherapy
Oestrogen Alkylating agents, radiotherapy to pelvis/TBI
Adrenals Cortisol High-dose steroids (mostly haematological malignancies)
64
LATE EFFECTS OF CANCER TREATMENT AND SURVIVORSHIP 6
CNS tumour. Given the recent change in treat- Mental health issues must not be underesti-
ment to reduce mediastinal radiotherapy (a major mated and addressing them throughout treat-
cause of these sequelae in Hodgkin’s disease) it ment will aid with patients seeking help if
is hoped that these will reduce in the future. necessary, even many years later. Recognizing
that this is a potential problem and consequence
Psychological impact of cancer of losses associated with cancer and its treatment
allows it to be seen as a normal process which,
diagnosis and treatment like the rest of oncology supportive care, should
Following a diagnosis with cancer patients and be appropriately managed.
their families make psychological adjustments to
cope with the treatment and potential outcome. Relationships
Stress is, not surprisingly, very common and
On average cancer survivors are less likely to be
occurs in over 95% of patients. The majority of
married and have a higher incidence of divorce
these will have anxiety and mild depression
than their peers.
which may not require treatment. However
some will have more significant psychological
symptoms and needs including depression, post- Social consequences
traumatic stress disorder, suicidal ideation and When compared with age matched controls
various psychoses. cancer survivors have a lower income. They are
In survivors of childhood cancer there is a more likely to have difficulties obtaining life and
greater degree of some aspects of psychological health insurance or a mortgage after surviving
distress in the parents than in the patient them- cancer.
selves. Parents were more concerned about their
child’s health and thought more often of the Further reading
cancer and its diagnosis than the patient.
Ganz PA. Cancer survivorship. Today and Tomorrow.
Suicidal ideation and previous attempts at Springer Press. 2007. ISBN-10: 0-387-34349-0.
suicide have been shown to be present in up to Mertens A, Yasui Y, Neglia J et al. Late mortality in
12.83% of childhood cancer survivors. Stand- Five-year survivors of childhood and adolescent cancer:
ardized mortality ratios for deaths as a result of The childhood cancer survivor study. J Clin Oncol
suicide in cancer patients are in the order of 2001;13:3163–3172.
Expert guidance on fertility: The effects of cancer treatment on
1.35–2.9 compared with the general population. reproductive functions. Guidance on management. Report
Risk factors include male sex, older age, higher of a Working Party. Available from: http://www.rcr.ac.uk/
disease stage, poor prognosis, poor performance publications.aspx?PageID=149&PublicationID=269.
status, alcoholism, other psychiatric illness,
fatigue, pain, loss of function and previous or Helpful websites
family history of suicide attempts. Lack of family The USA National Action Plan for Cancer Survivorship:
or social support also correlates with increased http://www.cdc.gov/cancer/survivorship/pdf/plan.pdf
suicide risk. SEER survival data: http://seer.cancer.gov/csr/1975_2005/
65
Palliative care
TV Ajithkumar
7
Introduction Management of pain
Palliative care is the active holistic care of patients The management of pain includes analgesics
with advanced progressive illness and includes as well as non-pharmacological measures. The
areas other than oncology. Apart from, managing World Health Organization (WHO) analgesic
pain and other symptoms, palliative care is aimed ladder (Figure 7.1) has been the gold standard in
at delivering psychological, social and spiritual the management of pain and has been shown to
support to patients and their family to achieve the eliminate pain in 80% of patients. The remaining
best quality of life. Development of cancer pallia- 20% have complex pain which may require spe-
tive care and hospices has gained momentum in cialist interventions. Measures used in complex
the UK since the opening of the World’s first pain include neuro-anaesthetic interventions,
hospice in London in 1967 by Cicely Saunders palliative surgery, radiotherapy, chemotherapy,
and colleagues. Today, palliative care and end-of- physiotherapy, occupational therapy, and psy-
life care are growing areas of research worldwide. chosocial care.
This chapter deals mainly with symptom manage- Analgesics
ment in cancer patients and a detailed discussion
of the principles of multidisciplinary palliative Commonly used analgesics are given in Table
care is beyond the scope of this chapter. 7.1. Strong opioids are started at a low dose and
titrated according to the clinical need (Box 7.2).
Morphine is the strong agent of choice and oral
Pain management administration is preferred. Transdermal prepa-
Pain control is an important component of cancer rations are useful only in stable pain. Some agents
management and uncontrollable pain is the major may only be prescribed by specialists in palliative
fear for many cancer patients. More than 80% of care medicine or anaesthetia.
patients with advanced cancer suffer pain and Opioid side effects, toxicity and management
around 20% of pain in cancer patients may be All patients feel some degree of drowsiness when
attributed to surgery, radiotherapy and chemo- starting or increasing their dosage. This usually
therapy. Pain control involves two important wears off 3–5 days after being on a stable dose.
steps: assessment of pain and management of pain. Constipation is inevitable and 30–50% patients
develop nausea and vomiting; these symptoms
Assessment of pain should be managed prophylactically. Intolerable
It is important to assess the multidimensional side effects necessitate switching to an alternative
nature of pain. Intensity of pain, location, dura- opioid (Figure 7.2).
tion and factors that modify pain should be Opioid toxicity usually manifests pseudo-
assessed. There are various tools to assess the hallucinations (shadows at the peripheries of the
pain and the commonly used ones are shown in field of vision), myoclonic jerks, cognitive impair-
Box 7.1. ment, and visual and auditory hallucinations.
66 © 2011, Elsevier Ltd
DOI: 10.1016/B978-0-7234-3458-0.00012-9
PALLIATIVE CARE 7
Box 7.1: Numerical rating score (NRS) – Wong-Baker FACES Pain Rating Scale.*
0–5 coding 0 1 2 3 4 5
0-10 coding 0 2 4 6 8 10
No hurt Hurts little Hurts little Hurts even Hurts whole Hurts worst
bit more more lot
*From Hockenberry MJ, Wilson D: Wong’s essentials of pediatric nursing, ed. 8, St. Louis, 2009, Mosby. Used with permission. Copyright Mosby.
Figure 7.1
Interventional measures
The WHO analgesic ladder. *NRS –
numberical rating score using 10
Step 3: strong opioids point coding.
First line: Morphine
Alternative: diamorphine, fentanyl
± step 1 ±adjuvant
± step 1 ±adjuvant
Initial management includes adequate hydration managed by reducing doses of opioids over a
(dehydration is often a precipitant of toxicity), few days.
reduction of opioid and haloperidol (1.5–3 mg
oral) for cognitive impairment. If the patient has Adjuvant analgesics
persistent toxicity without adequate analgesia, Adjuvant analgesics (Table 7.2) are a useful
opioid switching is needed (Figure 7.2). Though complement to regular analgesics in complex
there is no best choice, oxycodone and hydro- pain. These include anticonvulsants, antidepres-
morphone are the usual alternatives. Naloxone sants, antispasmodics, bisphosphonates, steroids,
is a short acting opioid antagonist used intrave- muscle relaxants and N-methyl-D-aspartate
nously for reversing accidental severe opioid antagonist (ketamine).
overdose.
If pain is relieved rapidly, for example after a Complex pain
nerve block, opioids can be stopped abruptly in Neuropathic pain is often difficult to control.
many patients without any side effects. However, Agents useful to control neuropathic pain are
up to 10% may experience a withdrawal syn- shown in Table 7.2. Amitryptyline along with
drome due to physiological dependence which is regular analgesics is the first line management. If
67
I BASIC PRINCIPLES OF CANCER MANAGEMENT
68
PALLIATIVE CARE 7
Oral morphine
mg/24 hour
Cannot take oral morphine Cannot tolerate morphine due to side effects
Divide by 10
Breakthrough analgesia
• Same opioid and same route – 1/6th of 24 hour dose
Subcutaneous alfentanil • Fentanyl patch – 1/5 patch dose as diamorphine mg dose
(for breakthrough analgesia and safe in renal failure) • Alfentanil – 1/10 of 24 hour
Figure 7.2
Conversion of opioids.
70
PALLIATIVE CARE 7
intestinal motility and colic (octreotide 600–
Table 7.3: Agent of choice in nausea and 800 mcg/day subcutaneously).
vomiting based on cause
Treatment – first line
Cause anti-emetic
Oral infections
Chemotherapy Granisetron 1 mg BD PO
Ondansetron 8 mg PO BD Mouth care is important in cancer patients.
Dexamethasone 4–8 mg Immunosuppression and steroids predispose to
OD PO oral candidiasis, which manifest as white-yellow
Metoclopramide 20 mg
QDS PO
plaques or as a painful bleeding red area. Treat-
ment is with anti-fungals, e.g. oral fluconazole
Radiotherapy Granisetron 1 mg BD PO and may be given prophylactically with high risk
Ondansetron 8 mg OD
Haloperidol 1.5 mg OD or
treatments. Herpes simplex virus presents as
BD PO painful, yellow lesions on the oral mucosa. Sys-
Raised intra-cranial Cyclizine 50 mg tds PO
temic anti-viral agents and strong opioid analge-
pressure sics are usually needed. Bacterial infections are
treated according to culture results.
Delayed gastric Metoclopramide 10–20 mg
emptying QDS PO
Domperidone 10–20 mg
QDS PO Hiccups
Drug induced Haloperidol 1.5 mg OD or Hiccups are a reflex spasm of the diaphragm
BD PO. causing sudden movement of the glottis. Common
Metabolic e.g. uraemia Haloperidol 1.5 mg OD or causes include gastric distension, raised intra-
or hypercalcaemia BD PO. abdominal pressure, high dose steroids and elec-
trolyte imbalance (e.g. hyponatraemia, uraemia).
Metoclopramide (10 mg qds PO) is useful in
gastric distension. Dimethicone which is an anti-
foaming agent which eases flatulence and disten-
sion (Asilone 5 ml qds) can be effective. Baclofen
Intestinal obstruction (5 mg tds PO) or nifedipine (5 mg tds) can act as
Intestinal obstruction is common with colorectal smooth muscle relaxants. When these measures
and ovarian cancers. Treatment depends on a fail, chlorpromazine can be used as a central
number of factors. Surgery is the treatment of hiccup reflex suppressant (12.5–25 mg daily).
choice if feasible; however, many patients are only
suitable for conservative management. Depend-
ing on the level of obstruction, surgical interven-
Breathlessness
tion includes bypass surgery, stent or surgical Breathlessness is the subjective experience of dis-
excision. In general surgery is only recommended comfort in breathing. Common causes include
if there is a single focus of tumour or adhesion disease affecting the lungs, increased intracranial
causing compression. Multiple areas of involve- pressure, anaemia and pulmonary embolism.
ment or extensive peritoneal involvement is best Supportive measures include breathing exercises,
managed by conservative measures. Conservative a stream of air, either from a fan or through an
management is by bowel rest, nasogastric tube (in open window, and treatment of infections,
case of persistent vomiting), parenteral steroids cardiac failure, effusion, COPD and anaemia.
(to control nausea, bowel oedema and extrinsic Radiotherapy is useful for large tumours and
compression, control of nausea and vomiting bronchial obstruction. Lymphangitis carcinoma-
(cyclizine when there is complete obstruction) tosis is treated with dexamethasone 16 mg daily.
and metoclopromide (in partial obstruction) and Oxygen therapy is useful in patients with SaO2
somatostatin analogues to reduce gastrointestinal <90%. Morphine 5 mg 6-hourly is also useful in
secretions, promote absorption of fluid and reduce breathlessness.
71
I BASIC PRINCIPLES OF CANCER MANAGEMENT
73
Head and neck cancer
AA Edwards and RL Mendes
8
Introduction HPV and about 90% of these are positive
for HPV-16. HPV-16 is particularly asso-
Head and neck oncology is challenging. It ciated with oropharyngeal SCC.
demands a thorough knowledge of head and • Epstein–Barr virus has been implicated in
neck anatomy to guide multi-modality treatment nasopharyngeal carcinoma. Plasma levels
and the treatment itself can lead to significant of EBV DNA before treatment and after
impairment in important functions such as swal- radiotherapy have been correlated with
lowing, facial expression and speech. Tumours outcome and survival.
arising within various head and neck subsites can • Other risk factors:
differ in presentation, clinical course and thera- • Lower socioeconomic status is associated
peutic options. with SCC of the oral cavity and larynx.
Cancers of the head and neck account for • Chewing betel quid – a mixture of tobacco,
around 6% of malignancies worldwide. It is slaked lime and areca nut, wrapped in
more common among men, with a male-female betel pepper leaf – is popular in India and
ratio of 2–3 : 1. Squamous head and neck cancers parts of South East Asia. It is associated
tend to affect patients between the ages of 40–70 the development of oral submucous fibro-
years. The incidence varies geographically – for sis and leucoplakia and is a risk factor
instance, nasopharyngeal carcinoma is most fre- for developing carcinoma of the oral
quent in the Far East. cavity.
• Occupational risk factors include: asbestos
Aetiology (larynx), wood dusts (nasal cavity, nasal
sinuses, nasopharynx and larynx), nickel
The important aetiological factors in squamous
(maxillary sinus) and pesticides (larynx).
cell carcinomas of the head and neck include:
• UV exposure is a risk factor for SCC of
• Tobacco – smoking or chewing tobacco is the the lip.
strongest risk factor for squamous cell carci- • Previous head and neck irradiation
nomas (SCC) of the head and neck, with more increases the risk of subsequent head and
than 90% of patients having a history of neck malignancies.
tobacco use.
• Alcohol – increased alcohol consumption is
associated with an increased risk of head and
Pathogenesis and pathology
neck SCC, and has been shown to have a SCC is thought to develop in a stepwise progres-
synergistic effect with tobacco in the develop- sion from squamous metaplasia to dysplasia,
ment of SCCs. through carcinoma-in-situ to invasive squamous
• Viral infections: cell carcinoma. These histological changes are
• Human papilloma virus – About a quarter accompanied by molecular alterations which
of head and neck SCC specimens contain disrupt the regulation of cell proliferation, by
inactivating tumour suppressor genes and acti- lymphatic drainage for head and neck subsites,
vating oncogenes. which are important in planning surgery and
The majority (90%) of malignant tumours of radiotherapy. In the unoperated neck, the pattern
the head and neck are squamous cell carcinomas. of lymph node drainage is relatively predictable
The WHO classification of head and neck cancers for different tumour subsites. The risk of occult
is outlined in Box 8.1. lymph node metastasis varies according to the
primary site and the size of the primary tumour.
Clinical assessment of this risk of cervical nodal
Anatomy metastasis dictates subsequent decisions on inclu-
Figure 8.1 shows the anatomical sites in the head sion of lymph node groups within a neck dissec-
and neck. Figure 8.2 demonstrates the anatomi- tion or radiotherapy target volume during the
cal levels of neck nodes and the typical regional definitive treatment.
Figure 8.2
IA – Submental Anatomical levels of neck nodes.
IB – Submandibular
VB – Supraclavicular
III VI – Anterior compartment
nodes
VA
VI
IV VB
78
HEAD AND NECK CANCER 8
A B
Figure 8.3
MRI of nasopharyngeal cancer.
Table 8.1: Risk factors for predicting locoregional recurrence in the postoperative setting
Primary site Neck nodes
Major risk factors Positive resection margins Extracapsular spread
Minor risk factors Close resection margins (<5 mm) 2 or more involved nodes
Invasion of soft tissues More than one lymph node level involved
Multifocal primary Involved node >3 cm in diameter
Perineural invasion
Vascular invasion
Poorly differentiated
T3 or T4 disease
High risk of recurrence is associated with the presence of one major risk factor or two minor risk factors.
Intermediate risk of recurrence is associated with the presence of one minor risk factor.
Factors of less importance in predicting risk of recurrence include: oral cavity primary site; presence of
carcinoma-in-situ or dysplasia at the resection margin; and uncertain surgical or pathological findings.
advanced head and neck cancer has been Role of biological agents
controversial. A large meta-analysis of trials Epidermal growth factor receptor (EGFR) inhib-
studying the role of chemotherapy in locally itors have been studied in head and neck cancer.
advanced head and neck SCC showed no signifi- Over 80% of head and neck SCC tumours
cant benefit for induction or adjuvant chemo- overexpress EGFR. This is associated with
therapy. More recent studies have supported a aggressive tumours, resistance to cytotoxic
role for induction chemotherapy in improving agents and irradiation, hence a poor prognosis.
laryngeal preservation in locally advanced SCC Cetuximab combined with radical radiotherapy
of the larynx. A study comparing cisplatin-5-FU is proven to improve both 3-year local control
with docetaxel, cisplatin and 5-FU showed a (41% vs. 34%) and overall survival (55% vs.
better survival with three drugs (Box 8.5). 45%) in locally advanced head and neck SCC
Palliative compared with radiotherapy alone. Hence NICE
recommends the use of cetuximab with radio-
Palliative chemotherapy provides a modest
therapy in patients with locally advanced head
benefit in advanced head and neck cancer with
and neck cancer with good performance status
objective response rates of 30–40% and median
in whom platinum-based chemoradiation is
survival around 6 months. Cisplatin/5-FU com-
contraindicated.
binations are most commonly used although
capecitabine, taxanes and biological targeting
agents are all being studied. Patients who fail to
respond to first line chemotherapy should be con-
Care during and after treatment
sidered for phase 2 experimental studies. Head Radiotherapy toxicity is discussed on p. 348.
and neck cancer provides an ideal template for Rehabilitation of the patient after completion
the study of novel agents as its accessibility of treatment involves input from a multi
permits scrutiny of both tumour control and disciplinary team including clinical nurse special-
normal tissue effects. The EXTREME study ists, speech and language therapists, dietitians,
showed that addition of cetuximab to cisplatin physiotherapists, occupational therapists, dental
and 5-FU improves overall survival compared to surgeons, dental hygienists and prosthetic
chemotherapy alone. specialists.
82
HEAD AND NECK CANCER 8
A B
Figure 8.5
An axial CT slice of the neck shows an IMRT plan with (A) concave isodose patterns around the spinal cord, sparing it
from the high-dose region and (B) another plan shows sparing of parotid.
84
HEAD AND NECK CANCER 8
Hypercalcaemia either due to excess parathyroid nodes. CT is not reliable in assessing lymphaden-
hormone (PTH) or PTH-related peptides is opathy, to distinguish T3 from T4 disease and in
common in squamous cell carcinomas (15%). demonstrating chest wall invasion, which all are
Hyponatremia occurs in up to 10% of patients, important in making treatment decision.
which is either due to syndrome of inappropriate CT scan is useful in assessing resectability.
secretion of ADH (SIADH) or to atrial natri Encasement of proximal pulmonary arteries/
uretic factor. SIADH is commonly associated veins, gross mediastinal involvement by tumour,
with SCLC. widespread mediastinal lymphadenopathy and
distant metastasis on CT scan suggest an unre-
Signs sectable tumour (96% accuracy). Tumour inva-
A normal physical examination does not exclude sion of central pulmonary artery and vein,
lung cancer. involvement of main stem bronchus and tumour
extension across the major fissure (anywhere on
General: cachexia, finger clubbing, anaemia,
the left side, above the minor fissure on right),
enlarged supraclavicular lymph node, features
all suggest the need for pneumonectomy for com-
of SVCO and Horner’s syndrome (in upper
plete resection.
lobe tumours).
CT scan of brain is done if there is clinical
Chest: reduction of breath sounds over a lobe,
suspicion of brain metastasis or patient is planned
signs of lobar collapse, inspiratory crackles
to undergo curative treatment.
over a lobe, unilateral wheeze, features of
Bone scan is indicated if there is bone pain or
pleural effusion, rib tenderness.
isolated raised alkaline phosphatase.
Other systems: bone tenderness, hepatomeg-
aly, features of brain metastasis, peripheral
neuropathy and proximal myopathy. Tissue diagnosis
Flexible bronchoscopic biopsy can be obtained
Investigations and staging (Figure 9.1) in 60–70% of lung cancers (central tumours).
Evaluation of a patient with suspected lung Sampling using multiple techniques (biopsy,
cancer is to: brushings and washings) gives the highest diag-
nostic yield (sensitivity of 83–88%).
• establish histologic type
CT guided percutaneous needle aspiration/
• define extent of disease (staging)
biopsy – is useful in peripheral tumours. Core
• assess fitness to undergo the best appropriate
biopsies improve the sensitivity compared with
treatment.
aspirates. Complications include bleeding and
Imaging pneumothorax.
Fine needle aspiration is suitable for sampling
Chest X-ray (Figure 9.2A) may show lung lesion
lymph nodes, skin nodules and liver and adrenal
with or without lymphadenopathy. There can be
metastases. Pleural effusion requires aspiration,
associated lung collapse, pleural effusion, syn-
biochemical analysis and cytology together with
chronous nodules or pericardial effusion. Bone
multiple pleural biopsies.
metastases or bony destruction due to direct
Sputum cytology has wide variation in sensi-
invasion can also been seen.
tivity (10–97%) and should be reserved for
Contrast enhanced CT scan of chest, includ-
patients with central tumours who are unable to
ing upper abdomen (3–10% patients show
tolerate or unwilling to undergo bronchoscopy
asymptomatic liver and/or adrenal metastasis –
or other invasive procedures.
Figure 9.2B&C) should be performed prior
to further diagnostic investigations including
bronchoscopy. Staging
Lymph nodes of >1 cm in short axis diameter, Stage determines prognosis and guides treat-
a central low intensity suggesting necrosis and ment. TNM staging of lung cancer is given in
rounding of the contour of a hilar node where it Box 9.3. Small cell lung cancer can also be staged
meets the lung margin suggest malignant lymph using a simple staging classification (Box 9.4).
92
CANCERS OF THE THORAX 9
Yes
PET scan
Definite distant metastasis or Suspicious of mediastinal disease Negative for mediastinal disease
high probability of N2/3 disease or distant metastasis and distant metastasis
Figure 9.1
Evaluation of lung cancer.
93
II SITE-SPECIFIC CANCER MANAGEMENT
A B
Figure 9.2
Imaging in lung cancer.
ing paratracheal (2 & 4), scalene and inferior and referral to a team specializing in the manage-
mediastinal nodes (7, 8 & 9). It is not useful in ment of lung cancer. Treatment depends on type
evaluating aortic nodes (5 & 6). Mortality rates of lung cancer (NSCLC vs. SCLC), stage, per-
from mediastinoscopy are negligible, but mor- formance status, co-morbidities and cardiopul-
bidity rates especially arrhythmias are reported monary reserve.
to be 0.5–1%. It has to be borne in mind that up
to 30% of patients with lung carcinoma who Non-small cell lung cancer (Figure 9.6)
have a negative mediastinoscopy prove to have Stage I–II
mediastinal nodal metastasis at surgery.
Video-assisted thoracoscopy (VATS) is useful Patients presenting with stage I–II disease are
to evaluate aortic (5 & 6), paraoesophageal (8) generally treated with curative intent if perform-
and pulmonary ligament nodes (9). It is also ance status is good (0–1) and pulmonary func-
useful in assessing the pleural cavity and obtain- tion are adequate. The treatment options include:
ing pleural biopsies. • Surgery alone (if excision complete).
• Surgery followed by radiotherapy (in case of
Management of lung cancer microscopic residual disease).
Patients presenting with symptoms suggestive of • Surgery followed by chemotherapy (in selected
lung cancer need urgent evaluation (Figure 9.1) cases of IB & II).
96
CANCERS OF THE THORAX 9
Excretion of isotope
through renal system
Figure 9.4
PET scan in lung cancer.
• Radical radiotherapy (in patients who are A complete anatomical resection is achieved
unfit or unwilling for surgery). by one of the following procedures:
Radical surgery • Lobectomy is the most common procedure
Surgery offers the best prospect of cure in stage which carries a postoperative mortality of
I–II disease, but only 10–20% of patients are suit- 2–4%.
able for surgery. Surgery is aimed at complete • Pneumonectomy is indicated if the tumour
resection of the tumour and its intrapulmonary invades hilar structures (e.g. main bronchi,
lymphatics. Patients need careful preoperative main pulmonary artery) and has a higher
evaluation as they are often frail and frequently mortality rate (6–8%).
have cardiopulmonary morbidity due to chronic • Wedge or segmental resection is useful in
smoking. Spirometry is required in all patients. If patients with peripheral tumours and impaired
the FEV1 is less than 1.5 L for lobectomy and less lung function but carries a high rate of local
than 2 L for pneumonectomy, full lung function recurrence (3–5 times) and a lower survival
tests are required. Table 9.1 shows minimal pul- (reduce survival by 5–10%).
monary function required for different curative • Bronchoplastic or ‘sleeve’ resection is designed
treatments. All patients require an ECG and to spare lung tissue as an alternative to pneu-
patients with murmurs should have echocardio- monectomy. This procedure is used to resect
gram. Patients with known ischaemic heart disease endobronchial lesions at or adjacent carina to
should have detailed cardiologic assessment. preserve distal tissue.
97
II SITE-SPECIFIC CANCER MANAGEMENT
A C
B D
Figure 9.5
Endobronchial ultrasound (A) and its use for FNA (B) of an enlarged lower paratracheal node (C&D).
Radical radiotherapy (Table 9.2) tumour localization and radiation delivery. Early
Patients with medically inoperable stage I/II are results suggest a 2-year local control of >90% in
treated with radical radiotherapy if lung function patients with stage I tumours with <5% signifi-
is adequate (Table 9.1). In the only randomized cant toxicity.
trial, radiotherapy was inferior to surgery (4-year
survival 7% vs. 23%). After conventional radical Postoperative treatment
radiotherapy (60 Gy in 30 fractions over 6 Radiotherapy
weeks), 2-year survival is about 20%. A UK There is no proven role for postoperative
MRC study showed that 2-year survival is better radiotherapy after complete surgical resection of
(20 vs. 29%) with continuous hyperfractionated stage I–II lung cancer. There is some suggestion
accelerated radiotherapy (CHART). This radio- that routine radiotherapy in N0–N1 disease sig-
therapy regime consists of 54 Gy in 36 fractions nificantly decrease survival. Hence radiotherapy
over 12 continuous days giving three fractions may be offered in incomplete resection and/or
per day with a minimum interval of 6 hours unexpected N2 disease (improve local control
between each fraction. with no effect on survival) if performance status
An evolving technique of stereotactic radio- and residual lung function permits. However,
therapy typically delivers high doses of radio- further studies are needed to define the exact role
therapy in 3–5 fractions using precise method of of radiotherapy in the adjuvant chemotherapy era.
98
CANCERS OF THE THORAX 9
Stage I and II Stage III A and Stage III B (dry) and • Stage III B (wet) and IV
WHO PS 0-1 WHO PS 0-1 • All stages with WHO PS
2 or more
Neoadjuvant Neoadjuvant
chemotherapy x 3 chemotherapy x 3
cycles cycles
Adequate cytoreduction
to treat with radical radiotherapy No
Fit for surgery
Options
Yes • Palliative chemotherapy*
• Biological agents
• Palliative radiotherapy
Radical • Supportive care
radiotherapy
Adequate cytoreduction No
Yes No Yes
Surgery or radical
radiotherapy
Recurrence
Figure 9.6
Management of non-small cell lung cancer. *palliative chemotherapy is considered only in patients with good
performance status (0-2).
99
II SITE-SPECIFIC CANCER MANAGEMENT
*For peripheral tumours and small tumours (planning volume <150 cm3) >0.7 L.
$
For small tumours >1 L.
*V20 (volume of normal lungs receiving 20 Gy) should be less than 34% and spinal cord dose should be <38–40 Gy with 4 weeks’
treatment and <44 Gy with 6 weeks’ treatment.
Chemotherapy (Box 9.5) tin. 5-year overall survival in NCIC JBR 10 trial
In randomized trials, there is no survival benefit was increased from 54 to 69% in stage IB and II
with adjuvant chemotherapy for stage IA. In disease. In the ANITA trial overall survival was
stage IB and II, an early meta-analysis showed a not significantly different in stage IB (62% vs.
5% overall survival benefit with postoperative 63%) but was statistically significant in stage II
cisplatin-based combination chemotherapy. This (52% vs. 39%) resected NSCLC. More hetero-
has been supported by several recent trials of geneous trials which have allowed a wider range
cisplatin combinations. The NCIC JBR 10 and of tumour stages, more variety in chemotherapy
ANITA trials both used vinorelbine and cispla- combinations within the trial and less stand
100
CANCERS OF THE THORAX 9
Stage IIIB with malignant effusion/stage IV which cannot be encompassed in a radical radio-
Patients with malignant effusions (IIIB) or meta- therapy field or with co-morbidity. An MRC
static disease (IV) are incurable and both carry study showed that for such patients with per-
a similar prognosis. The treatment aim is pallia- formance status 0–1, high-dose palliative radio-
tive with the intention of improving symptoms therapy (39 Gy in 13 fractions, if no spinal cord
as much as disease control. Treatments will in the field or 36 Gy in 12 fractions, if cord in
be greatly affected by patient fitness as these the radiotherapy field) results in better 2-year
patients will often be frail and suffer several co- survival (13% vs. 9%) compared with 17 Gy in
morbidities. Many trials have examined the role two fractions. Patients who need relief of local
of chemotherapy versus best supportive care in symptoms due to primary tumour or metastases
this group. Overall chemotherapy has a response are treated with palliative radiotherapy. This is
rate of 20–40% with a duration of 6 months and either 10 Gy single fraction (PS >2), or 17 Gy in
a median overall survival of less than 1 year. The two fractions (PS 2). The side effects of large
survival benefit from chemotherapy is in the fractions include nausea, acute chest pain and
range of 6–8 weeks. Response rates for an indi- fever.
vidual reflected overall survival in a meta-analy-
sis. Frequent assessment prior to chemotherapy Relapsed pre-treated NSCLC
with CXR is recommended to assess response. In the small number of stage IIIB/IV patients who
Chemotherapy has been associated with sym have received previous platinum-based chemo-
ptomatic benefits such as reduced palliative therapy and relapse whilst still fit, second line
radiotherapy requirements, reduced analgesia, chemotherapy can be considered. Docetaxel has
weight stabilization and maintenance of per- shown good activity and improved median sur-
formance status. Toxicity was frequently seen, vival of 3 months over best supportive care in
worse with combination chemotherapy but this subset of patients. Re-irradiation is an option
quality of life studies showed that these were not if >6 months after previous radiotherapy and it
perceived as worse compared with patients not is possible to avoid spinal cord.
on chemotherapy. Erlotinib has shown a survival advantage of 2
In general, chemotherapy has a greater benefit months and improved symptom control when
in fit patients of good performance status (WHO used in combination with chemotherapy. In the
0 or 1). In these patients combination chemo- UK, erlotinib is recommended as a clinically and
therapy (a platinum and other agent such as cost-effective alternative to docetaxel for the
vinorelbine or gemcitabine) can be offered. A second line treatment of NSCLC.
recent study has shown that a combination of
cisplatin and pemetrexed improves overall sur- New agents
vival in patients with locally advanced or meta- Understanding some of the molecular mecha-
static adenocarcinoma (12.6 vs. 10.9 months) nisms in NSCLC has led to the development of
and large cell carcinoma (10.4 vs. 6.7 months) new targeted therapies. The mechanisms of these
compared with cisplatin and gemcitabine. In the are common to several malignancies (see
less fit or elderly single agent treatments, pallia- Appendis, p. 370). The most successful agents in
tive radiotherapy, biological agents or appropri- NSCLC are those targeted against the epidermal
ate trials can be considered. growth factor receptor (EGFR). Two oral drugs
Recently phase III studies have shown that which inhibit the tyrosine kinase domain of the
maintenance treatment with pemetrexed and EGFR are erlotinib (Tarceva™) and gefitinib
erlotinib are useful in patients who had not pro- (Iressa™). Toxicity with these drugs is unlike
gressed after first line chemotherapy. standard chemotherapy drugs and includes a
characteristic rash which often reflects drug
Palliative radiotherapy activity, diarrhoea and pneumonitis (reported in
A significant number of patients may need radio- gefitinib in a Japanese study). The patients most
therapy to palliate symptoms. There will be a likely to benefit from these drugs are female,
subgroup of patients with large localized NCSLC those with an adenocarcinoma (especially bron-
102
CANCERS OF THE THORAX 9
Palliative chemotherapy
Performance status and/or radiotherapy Performance status
No No
0–2 • 0–2
Supportive care
Yes Yes
No Yes
Figure 9.7
Management of small cell lung cancer.
choalvelolar subtype), be non-smokers and be Small cell lung cancer (Figure 9.7)
Asian. At the molecular level, increased EGFR Small cell lung cancer (SCLC) accounts for 20%
protein expression, gene amplification and the of lung cancer. Up to 60% have extensive stage
presence of EGFR mutations in the tyrosine disease at diagnosis.
kinase domain increase patient response. A recent
comparative study of gefitinib with a combina- Limited stage disease
tion of carboplatin and paclitaxel (IPASS study) The standard treatment for patients with good
has shown that gefitinib results in a better pro- performance status (0–2) and no significant co-
gression free survival in patients with EGFR+ morbidity is combination chemotherapy with
tumours. platinum regime with concurrent thoracic radio-
A randomized study (E4599) showed that a therapy and prophylactic cranial radiotherapy.
combination of bevacizumab (anti-VEGF) with In 5–10% of patients with SCLC limited to
chemotherapy improves survival by 2.3 months lung, diagnosis is established only after resection.
in patients with untreated advanced non-squa- These patients need adjuvant chemotherapy as
mous cell lung cancer. there is high likelihood of development of distant
103
II SITE-SPECIFIC CANCER MANAGEMENT
metastasis. Role of surgery in early stage disease therapy. Some others use a radiotherapy regime
is not well established. Surgery following com- of 50 Gy in 25 fractions. One study shows that
plete or partial response to chemotherapy is not twice-daily concurrent radiotherapy is better
beneficial. than once daily radiotherapy.
poor performance status or metastatic disease are tion (conflicting results). Patients with meta-
treated with palliative measures. chronous brain recurrence after a curative
resection are also considered for resection ±
Lung cancer in never smokers
WBRT or radiosurgery.
Never smokers are those who have smoked less
than 100 cigarettes in their lifetime. Studies
suggest that environmental tobacco smoke is Palliative care issues
a common cause of these cancers. Other risk Despite recent advances in treatment, many
factors may also contribute. Women are more patients with SCLC and NSCLC will progress
commonly affected than men. Adenocarcinoma and eventually die of their disease. Patients with
is more common, particularly the bronchoalveo- advanced disease can present with a number of
lar carcinoma subtype. Treatment is essentially problems which require specialist services.
the same as that of smokers. However, recent • Bone metastases are very common and can
data suggest that this group of patients have a result in pain and spinal cord compression (p.
higher response rate and better survival with 328). Pain can be improved using radiother-
EGFR tyrosine kinase inhibitors such as erlotinib apy with a single 8 Gy fraction or 20 Gy in
and gefinib (see IPASS study, p. 102). five fractions for spinal cord compression.
Multiple primary lung cancer • Haemoptysis: invasion of vessels by expanding
2–4% patients can present with multiple primary tumours can result in haemoptysis which can
lung tumour which can be either synchronous be significant. Palliative radiotherapy (10 Gy
(detected simultaneously) or metachronous single fraction) may alleviate the bleeding but
(detected after an interval). However it is often other measures can be used, e.g. cryotherapy.
difficult to distinguish this from a metastasis from • Superior vena cava obstruction can occur and
the lung tumour. Patients with multiple primary should be treated as an emergency (p. 337).
cancers are managed as two individual tumours. Steroids should be given in the first instance
and radiotherapy (20 Gy in five fractions) is
NSCLC with solitary metastases successful at reducing symptoms in 60%.
A small subset of patients present with isolated • Hypercalcaemia can be due to bone metas-
extrathoracic metastasis either after radical treat- tases or the production of a PTH like peptide.
ment or at presentation. Studies show that in It should be tested for regularly in advanced
patients with good performance status, resection lung cancer to prevent it presenting as
of the lung tumour and metastasis with or without an emergency (p. 339). It is treated with
systemic treatment result in improved survival. hydration, bisphosphonates and treatment of
In patients with synchronous isolated adrenal the underlying cancer if that is possible.
metastases, this approach results in a 5-year sur- • Stridor is a rare but distressing symptom
vival of 10–23%, whereas in those with isolated caused by compression of the proximal
metachronous adrenal metastasis occurring >6 airways, usually by tumour or lymph nodes.
months after initial curative treatment, the 5-year Initial supportive care includes assessment of
survival is around 38%. Of those with recurrence the airway by otolaryngologist, steroids and
in <6 months, none survive for 2 years. oxygen (p. 339). If severe, heliox, a mixture
In patients with synchronous isolated brain of helium and oxygen where the small particle
metastases with complete resection of all disease, size of helium facilitates delivery of the
the reported 5-year survival is 21%. Hence syn- oxygen, can be given. Treatment of the com-
chronous brain metastasis in a fully staged pressive lesion (e.g. radiotherapy to the nodal
(including PET scan) resectable stage I–II lung mass) may be the only option to prevent pro-
cancer patient with good performance status is gression of stridor.
treated with resection or radiosurgical ablation • Breathlessness is the most common symptom
with complete excision of the primary tumour. in advanced NSCLC and can be debilitating.
They also need systemic treatment as well as Investigations should be done to rule out
whole brain radiotherapy (WBRT) after resec- significant pleural effusion and pulmonary
106
CANCERS OF THE THORAX 9
emboli as these are both treatable. Assuming lioma. However, smoking combined with asbes-
the breathlessness is due to tumour progres- tos exposure increases the risk of lung cancer.
sion and no further active treatment is pos-
sible, simple measures such as the use of a fan, Pathogenesis and pathology
teaching relaxation techniques and the cau- Pathogenesis
tious use of oral morphine, which causes dila- Amphiobile (blue and brown) asbestos is strongly
tation of smooth muscle walls in blood vessels linked to mesothelioma. It is postulated that the
leading to an improvement in symptoms, can thin and long fibres of amphiboles when inhaled
be helpful. Overall lung cancer patients often could penetrate and scratch the pleural surfaces.
have an array of palliative care needs such The chromosomal changes caused by fibres as they
that specialist in symptom control should be pierce the mitotic spindle as well as free radical
involved at an early stage. formation from the fibres cause malignancy. The
Follow-up possibility of relationship between simian virus 40
(SV40) and mesothelioma is still debatable. Recent
Role of follow-up in patients after curative treat- studies suggest a possible genetic susceptibility to
ment for NSCLC remains controversial. For asbestos-induced carcinogenesis.
patients with potentially curative re-treatment,
it is reasonable to cover routine follow-up 3–6 Pathology
monthly for 2 years and 6–12 monthly there There are four histological subtypes of meso
after, with clinical examination and radiological thelioma:
evaluation. The role of routine follow-up after
treatment for SCLC is less clear. • Epithelial (40%) – associated with pleural
Some evidence suggests that continued smok effusions and better prognosis.
ing adversely affects the overall prognosis and • Sarcomatoid (20%) – ‘dry’ mesothelioma.
hence discouraged. • Mixed (35%).
• Undifferentiated (5%).
Mesothelioma The important histopathological diagnosis is
adenocarcinoma, which is differentiated immu-
Introduction nohistochemically. Mesothelioma is positive for
Mesothelioma is an aggressive tumour arising calretinin, WT1 (Wilms’ tumour 1) antigen, epi-
from the cells of pleura and peritoneum. Cur- thelial membrane antigen (EMA) and cytokeratin
rently just over 2000 new cases are diagnosed in 5/6 and negative for cytokeratin 7, 8, 18 & 19,
the UK annually and the number of cases is TTF-1, BER-EP4 (mostly), CEA and MOC. The
expected to peak in 2015. In the USA, 2000 new converse is diagnostic of adenocarcinoma.
cases are diagnosed annually and the incidence Presentation
is declining. Widespread use of asbestos after
World War II correlates with the anticipated The usual presentations are exertional dyspnoea,
increase in Europe within the next 15 years. The chest pain or pleural effusion. Weight loss, fatigue,
median age at diagnosis is 60 years and males are cough and paraneoplastic fever are also common.
five times more commonly affected than females. Right hemithorax (65%) is more often affected
than left (35%) and 3% cases are bilateral.
Aetiology
Investigations and staging
Mesothelioma is the most strongly linked cancer
to occupation. It is estimated that 80–90% of Imaging
mesothelioma cases or deaths in men are caused Chest X-ray and CT scan (Figure 9.8) are the
by occupational exposure to asbestos. The latency initial methods of imaging. Chest X-ray shows
period after exposure ranges from 15–50 years. evidence of pleural thickening, pleural effusion
Shipbuilding and construction are the industries and volume loss. CT scan shows unilateral
which used to involve high exposure of asbestos. pleural effusion, nodular pleural thickening and
Smoking does not increase the risk of mesothe- tumour encasement of the lung (‘rind like’
107
II SITE-SPECIFIC CANCER MANAGEMENT
Staging
The recommended staging system is the Inter
national Mesothelioma Interest Group (IMIG)
staging system, which is based on clinico-patho-
logical tumour variables and nodal classification
that of lung cancer (Table 9.3).
Management
There is no standard of care and only a minority
of patients (10–15%) are eligible for any poten-
tially curative treatment. The majority of patients
(85–90%) presents with advanced disease with
medial survival of less than 12 months and 5-year
A survival of ≤1%. Suitable patients with stage I
and II disease may be treated with radical surgery
or multimodality treatment aimed at long-term
survival.
Surgery
There are no randomized data on the benefits of
surgery. Radical treatment involves resection of
the entire parietal and visceral pleura en bloc
with underlying lung, ipsilateral pericardium
and diaphragm called extrapleural pneumon
ectomy (EPP). Historical series suggest 5-year
survival of 10–20% with EPP. The MARS trial
(Mesothelioma and Radical Surgery) is aimed to
B
determine whether EPP is better than no surgery
Figure 9.8 in terms of survival and quality of life. However
Imaging of mesothelioma. the majority of patients are not suitable for such
extensive surgery. It has a reported morbidity
appearance – seen in 70% of cases) leading to up to 60% and mortality of 3.4% in expert
contraction of hemithorax. Calcified plaques are hands.
found in 20% of patients. Chest wall invasion Palliative surgery involves pleurectomy or
leads to obliteration of extrapleural fat planes, decortication. It is an option for palliation of
invasion of intercostal muscles, displacement of breathlessness caused by effusion when standard
ribs, or bone destruction. treatment options failed. The exact benefits of
PET scan is helpful in distinguishing benign these procedures are not well-defined.
from malignant pleural masses and to detect Radiotherapy
extrathoracic and nodal disease. Recent studies
report increasing usefulness of PET scan in defin- Mesothelioma is not a radiosensitive tumour.
ing prognostic group and to assess response to However, radiotherapy is still used in the follow-
chemotherapy. ing settings:
• Prophylaxis of thoracic tract seeding – to
Tissue diagnosis avoid mesothelial cell seeding down the tracts
Histopathological diagnosis should be made in after thoracoscopies and drains. There is no
all cases using cytological analysis of pleural fluid convincing evidence for this indication (three
or pleural biopsy done either under radiological small trials; one showed benefit and two did
guided or by VATS. A cell block prepared from not). Radiotherapy dose is 21 Gy/3 fractions
108 at least 500 ml of pleural fluid is often helpful. over a week using electrons.
CANCERS OF THE THORAX 9
Chemotherapy
Table 9.3: IMIG staging system
for mesothelioma Although chemotherapy is used a part of multi-
modality treatment, many regimes have poor
Stage
response to treatment and severe toxicity. Many
Ia
chemotherapy studies using single or combina-
T1aN0M0 Limited to parietal pleura; no
involvement of visceral
tion regimens showed an objective response rate
pleura. of <20% with no impact on survival. A system-
Ib atic study suggested cisplatin as the most active
T1bN0M0 Limited to parietal pleura: single agent. The antifolate agent, pemetrexed,
scattered foci involving which inhibits enzyme thymidylate synthetase,
visceral pleura. represents the recent advance in chemotherapy.
II A recent randomized study of 456 patients with
T2N0M0 Ipsilateral pleura with (i) PS 0–1, cisplatin with pemetrexed vs. cisplatin
involvement of diaphragmatic alone showed a superior response rate (41.3 vs.
muscle or (ii) confluent
16.7%; p < 0.001), median survival (12.1 vs. 9.3
visceral pleural tumor or
extension from visceral pleura months; p = 0.02), and time to progression (5.7
into the lung parenchyma. vs. 3.9 months; p = 0.001). Neutropenia was the
III
commonest toxicity in the combined arm. The
T3N0M0 Ipsilateral pleura with recommended dose of pemetrexed is 500 mg/m2
involvement of the given IV on day 1 followed by cisplatin 75 mg/
endothoracic fascia, or m2 of each 21 day cycle for 4–6 cycles. There are
extension into mediastinal no data on benefit of chemotherapy beyond six
fat or solitary resectable
extension of chest wall or
courses.
non transmural involvement In elderly patients, carboplatin is often substi-
of pericardium. tuted for cisplatin and data from phase II studies
T1–3N1M0 Ipsilateral bronchopulmonary suggest similar efficacy.
or hilar nodes.
T1–3N2M0 Ipsilateral mediastinal or
internal mammary nodes/
Palliative care
subcarinal nodes. Recurrent pleural effusion is a debilitating
IV problem. Talc pleurodesis seems to give the best
T4N0M0 Diffuse or multiple chest results with low morbidity. Alternate option is
wall involvement; PleurX catheters with repeated drains.
transdiaphragmatic extension Management of other symptoms follows the
to peritoneum; direct
extension to contralateral
same principle of any advanced cancer.
pleura; mediastinal organ
extension; extension to spine; Prognostic factor and survival
extension to internal surface
There are two prognostic predictive models:
of pericardium or involvement
of myocardium. EORTC and Cancer and Leukaemia group-B
Any T, N3M0 Contralateral mediastinal (CAL-B). EORTC system (Table 9.4) distin-
nodes; contralateral internal guishes patients with mesothelioma as good
mammary nodes or any (median survival 10.8 months) and poor (5.5
supraclavicular nodes.
months) prognostic groups, where as CAL-B
Any T, Any N, M1 Distant metastases present.
distinguishes six groups with median survival
ranging from 1.4 months to 13.9 months. Table
9.4 shows the EORTC prognostic index.
• Palliative radiotherapy –to relieve pain.
• Adjuvant radiotherapy following EPP – no Medicolegal implications
randomized trial data. One phase II trial Mesothelioma is a notifiable disease. Patients
reported reduced local recurrence and pro- diagnosed to have mesothelioma are automati-
longed survival. cally eligible for some benefits and allowances. 109
II SITE-SPECIFIC CANCER MANAGEMENT
A B
Figure 9.9
Imaging in thymoma.
Thymoma – preoperative
• Adjuvant radiotherapy Consider RT if area of risk of recurrence Stage IVa – a good response
• If IB, risk of pleural disease – can be encompassed in a radical to chemotherapy – consider
consider adjuvant chemotherapy radiotherapy portal surgery ± radiotherapy
for young or fit patients
Relapse
Figure 9.10
Management of thymoma.
114
Breast cancer
TV Ajithkumar and HM Hatcher
10
Introduction Pathogenesis and pathology
Breast cancer is the most common female malig- Breast cancer arises from the epithelial cells lining
nancy in the UK and USA. In the UK, 30,000 the terminal duct lobular unit. Development of
new cases and 15,000 deaths occur each year due invasive breast cancer is thought to be due to a
to breast cancer. In the USA, there are 192,000 multi-step process. WHO classification of breast
new cases and 43,300 breast cancer deaths every cancer is shown in Box 10.1.
year. The life-time risk of developing breast
cancer for a woman is 1 in 12 in the UK and 1 Precursor lesions
in 8 in the USA. Lobular carcinoma-in-situ is considered as a pre-
cursor of invasive lobular cancer, but in most
cases, it does not progress to the invasive stage.
Aetiology It is regarded as a marker for either lobular or
The reported risk factors include hormonal, ductal carcinoma.
genetic, dietetic and radiation. Table 10.1 shows Atypical ductal hyperplasia is characterized by
these risk factors and protective factors for breast intraductal epithelial proliferation with some of
cancer. The effect of hormones, notably oestro- the features of DCIS. These increase the risk of
gen, is the most significant aetiological factor in invasive cancer 4–5 times.
breast cancer. Genetic factors in breast cancers In ductal carcinoma-in-situ (DCIS) malignant
are dealt with in Chapter 5 (p. 47). BRCA1 epithelial proliferation is confined to a duct with
mutation associated cancers tend to occur at an no stromal invasion. It accounts for 3–5% of
early age, are highly aggressive and are typically palpable breast cancer and 15–20% of screen
negative for oestrogen (ER), progesterone (PgR) detected cancer. It is graded according to the
and human epithelial growth factor receptor 2 appearance of cell nuclei. Low-grade micropapil-
(HER2/neu (’Triple negative’). BRCA2 muta- lary DCIS can occur as a multicentric tumour
tions account for 1% of breast cancers which are whereas high-grade comedo carcinoma DCIS can
often ER and PgR positive. be associated with invasive tumour in the same
Other risk factors include: quadrant.
Microinvasive carcinoma is focus of invasive
• Previous breast cancer – increases the risk of cancer of <1 mm in maximum extent.
a contralateral breast cancer (0.5–1% per
year). Malignant breast lesions
• Prior radiation – to the breast or part of • Invasive ductal carcinoma: 70–80% of all
breast increases the risk of cancer (RR 3) (e.g. invasive carcinomas are classified as invasive
Mantle radiotherapy for Hodgkin’s disease). ductal carcinomas, which are thought to arise
• Some benign conditions of the breast (e.g. from DCIS. They occur most commonly as
atypical hyperplasia). ‘ductal no special type’ (ductal NST). Ductal
Initial assessment
Figure 10.1 ‘Triple’ assessment of patients with suspected
Surgical anatomy of breast and lymphatic drainage. breast cancer includes a combination of clinical
117
II SITE-SPECIFIC CANCER MANAGEMENT
A B
A B
Figure 10.3
Ultrasound in breast cancer.
40 years with a high risk of breast cancer, to rule lesions may necessitate image localization using
out multifocal disease prior to conservation, mammography or ultrasound and hook wire
imaging women with implants and to distinguish replacement before open biopsy.
scars from tumour recurrence.
Pathological diagnosis
Investigations after
Pathological diagnosis may be obtained by core
triple assessment
biopsy or open surgical biopsy. Core biopsy can Most patients with early stage disease without
be obtained by conventional or vacuum assisted clinical evidence of metastatic disease will proceed
means such as Mammotome. Vacuum assisted to surgery after preoperative assessment. Patients
core biopsy yields a larger sample and may in who require neoadjuvant systemic treatment
some cases allow complete removal of the target prior to definite surgical management need
abnormality in the breast. Clinically impalpable staging investigations including:
119
II SITE-SPECIFIC CANCER MANAGEMENT
All of the following: Any of the following: 1–3 nodes positive 4 or more nodes positive
• pT ≤2 cm • pT >2 cm
• Grade 1 • Grade 2/3
• Vascular invasion • Vascular invasion
negative positive HER 2 negative HER 2 positive
• HER2 negative • HER2 positive
• Age ≥35 years • Age <35 years
Endocrine responsiveness
• Adjuvant chemotherapy for intermediate and high risk should be combination regimen with an anthracycline for a duration
of at least 4 cycles
• Taxanes should be considered for high-risk patients, especially ER negative
• Herceptin should be considered in patients with >1 cm HER2 positive tumour who had adjuvant anthracycline based chemotherapy
* Uncertain – low level of steroid hormone receptor immunoreactivity (<10% cells positive), lack of PgR, HER2 overexpression and
possibly µPA and PAI-1
HT – hormone therapy; CT – chemotherapy; OS – ovarian ablation; AI – aromatase inhibitor; Tam – tamoxifen;
GnRH - gonadotropin releasing hormone agonist
Figure 10.5
Adjuvant systemic treatment in breast cancer (based on St. Galen International consensus 2007).
122
BREAST CANCER 10
For SNB the sentinel node is localized by pre- Box 10.5: Radiotherapy in breast cancer
operative intradermal injection of technetium
Indications
labelled colloid and lymphoscintiscan or by
Whole breast
intraoperative injection of blue due or using both • After breast conservation for invasive cancer
techniques. Sentinel node biopsy has >95% • After breast conservation for DCIS (except low risk
detection rate. of recurrence)
Patients who are sentinel node positive will Tumour bed boost
undergo axillary clearance. A current study • All patients <40 years
(ALMANAC) is evaluating the role of axillary • 40–50 years with high risk features
radiotherapy in patients who are sentinel node • Patients with <1 mm margin and further surgery is
positive. not contemplated
Postmastectomy chest wall
Adjuvant radiotherapy • T3/T4 tumour
(Boxes 10.5 and 10.6) • ≥4 positive nodes after dissection
effects). All patients need bone mineral density therapy which is aimed at down staging the
assessment by DEXA scan prior to starting AI. primary tumour which may facilitate surgery or
Patients with osteopenia need vitamin D and even avoid the need for mastectomy. Full staging
calcium supplement whereas those with a T-score workup is needed in this group of patients.
less than −2.5 SD (osteoporosis) need bisphos- In older women with receptor positive
phonates. All patients on AI need a 2-yearly tumours, initial hormonal treatment can be used.
DEXA scan. Although studies show that all the AIs result in
a higher response than tamoxifen, letrozole is the
Adjuvant trastuzumab
only licensed drug for neoadjuvant use. Two ran-
In patients with HER2+ disease (immunohisto- domized studies, separately comparing letrozole
chemical score of 3+ or FISH score >2.0) one and anastrazole with tamoxifen, showed that
year of adjuvant trastuzumab is recommended. aromatase inhibitors induce a higher rate of
Trastuzumab (Herceptin) is a humanized mono- regression allowing breast conservation. In the
clonal antibody again the extracellular domain comparative study of 4 months of tamoxifen
of HER2. All the six randomized studies have with letrozole, letrozole has a higher response
shown improved DFS with addition of trastuzu- rate (55%) than tamoxifen (36%). This study
mab to chemotherapy while 4 of the 6 trials also showed that those patients with HER-1
showed an overall survival benefit with adjuvant or HER-2+ tumours had a higher response to
trastuzumab, e.g. HERA study which compared letrozole (88% vs. 21%). In the second study
chemotherapy vs. chemotherapy with trastuzu- anastrazole achieved significantly higher breast-
mab showed a better 3-year DFS (81% vs. 74%) conservation rate than tamoxifen (47% vs. 22%,
and OS (92% vs. 90%). p = 0.03).
Adjuvant trastuzumab is indicated in patients Neoadjuvant chemotherapy is the standard
with >1 cm and/or node positive tumour and treatment for patients with large primary tumours
who received adjuvant chemotherapy. The role aiming for conservation and for inflammatory
of trastuzumab in <1 cm and node negative breast cancer (see p. 132). Chemotherapy results
disease as well as a single modality is not studied. in a 70–90% response with a 20% pathological
A loading dose of 8 mg/kg followed by 17 complete response. NSABP B-27 showed that an
maintenance doses of 6 mg/kg every three weeks initial anthracycline regime followed by docetaxel
is the recommended schedule. If there are more results in a better response rate (90.7% vs.
than 7 days delay treatment needs to be restarted 85.5%, p < 0.001) and pathological complete
with a loading dose. response (64% vs. 40%, p < 0.001). In patients
Common side effects include hypersensitivity with HER2 positive tumours addition of trastu-
and flu-like symptoms. Cardiotoxicity is a zumab results in improved pathological response
concern (<4% symptomatic or severe cardiac (65% vs. 26%, p = 0.016) and it is advisable to
failure). Hence it is not given if the left ventricu- incorporate trastuzumab with the neoadjuvant
lar ejection fraction is <50%. It is not given con- non-anthracycline chemotherapy. One such
currently with anthracyclines, but can be given approach is to give initial 3–4 courses of anthra-
concurrently with taxanes. Cardiac monitoring cycline chemotherapy followed by taxanes with
during treatment is mandatory. A recent study trastuzumab. In breast cancer, type and degree
reported no increase in adverse cardiac events in of response to primary systemic treatment pre-
patients receiving trastuzumab with adjuvant dicts disease-free as well as overall survival.
radiotherapy. After neoadjuvant chemotherapy, inflamma-
tory breast cancer patients undergo total mastec-
Locally advanced disease tomy whereas non-inflammatory breast cancer
(T3 N1, T1–3 N2–3, T4 N0–3) patients undergo mastectomy or conservation if
appropriate. If patients had negative SNB pre-
(Figure 10.6) chemotherapy, no axillary surgery is needed. If
Patients with locally advanced breast cancer axillary nodal status is unknown or positive
are usually treated with neoadjuvant systemic SNB, axillary clearance is needed.
128
BREAST CANCER 10
Young women or ER – or fixed axillary nodes, Older women, ER+, non inflammatory Hormone negative/life-threatening
or inflammatory or aiming for conservation and no intent for conservation visceral involvement/failed hormones
Figure 10.6
Management of locally advanced and metastatic breast cancer.
All patients need postoperative radiotherapy Evaluation includes complete history and
to chest wall or breast (Figure 10.6). Adjuvant examination, blood tests including the tumour
systemic treatment depends on hormone receptor marker CA15-3 if available, CT scan chest,
and HER2 status. abdomen and pelvis, bone scan and receptor
status. Imaging of the brain is indicated only if
Metastatic disease symptomatic.
The aims of treatment are symptom control,
(any T, any N, M1) (Figure 10.6) improving quality of life and survival. Treatment
Approximately 6% patients present with distant options include hormones, chemotherapy and
metastasis at diagnosis and 40% of patients biological agents.
with early stage disease will eventually develop Patients with hormone receptor positive
metastases. The survival of patients with meta- disease without life-threatening visceral involve-
static disease varies from months to years. Bone ment are treated with hormonal treatment. In
metastases have the best outcome, followed by premenopausal women who have not previously
lung, liver and brain metastases. had tamoxifen or discontinued for >12 months,
129
II SITE-SPECIFIC CANCER MANAGEMENT
the standard option is tamoxifen and ovarian significant improvement in time to progression
ablation or suppression. Otherwise, an AI with with capecitabine combined with lapatinib com-
ovarian ablation may be considered. pared with capecitabine alone (6.2 months vs.
In postmenopausal women studies show that 4.3 months, p = 0.00013).
AI have superior results compared with tamoxifen A very small subset of patients present with
so should be the drug of choice, if not received bone marrow failure subsequent to extensive
previously. In one study, letrozole resulted in a bone disease. Modified chemotherapy with low-
significantly longer time to progression (41 vs. 26 dose weekly anthracycline or taxanes is the
weeks, p = 0.0001) and higher response rate (30% option in these patients (Box 10.6).
vs. 20%, p = 0.0006) compared with tamoxifen In patients with metastatic disease, response to
in advanced breast cancer. Since data are in more treatment should be evaluated after 3 months of
favour of letrozole, it is the agent of choice. There endocrine treatment and after 2–3 cycles of
is no definite recommendation for second-line, but chemotherapy.
the options include tamoxifen, anastrazole, letro-
zole, exemestane, fulvestrant and megesterol Recurrent disease
acetate. Unless there is an indication for chemo-
2–20% patients after conservative surgery and
therapy, various hormonal agents are used as
<10% patients after radical surgery will present
a cascade of treatment until all options are
with loco-regional recurrence within 10 years.
exhausted.
10–70% patients present with metastatic recur-
Chemotherapy is used in patients with recep-
rence within 10 years.
tor negative tumours, hormone resistant tumours,
Patients presenting with isolated loco-regional
and life-threatening progressive disease. Chemo-
recurrence need complete staging prior to poten-
therapy results in a 50% response rate usually
tially curative treatment and ‘secondary’ adju-
lasting less than one year. Based on meta-analy-
vant treatment. Patients with systemic recurrence
sis, anthracyclines result in a greater clinical
are treated similarly to those with metastatic
benefit than a CMF regime and should be
disease.
considered as first-line treatment if the patient
has not previously received an anthracycline.
Taxanes are the options after anthracycline
Palliative care
failure. Both docetaxel and paclitaxel have • Bone metastases – treated with pain manage-
proven activity. Other agents used in breast ment as per WHO analgesic ladder (p. 66)
cancer include capecitabine, vinorelbine, gemcit- and local radiotherapy studies show that
abine, carboplatin, etc. (Box 10.6). 8–10 Gy as a single fraction has similar effi-
Patients with HER2 positive disease are cacy to that of prolonged. Women with bone
treated with trastuzumab with or without chem- metastases should be given bisphosphonates
otherapy. In patients who progress on trastuzu- which help to control pain as well as prevent
mab, retrospective data support a change of fractures, need for radiotherapy or orthopae-
chemotherapy with continuation of trastuzu- dic surgery and epidoses of hypercalcaemia.
mab; however, NICE recommends discontinua- Pamidronate, ibandronate, clodronate and
tion if it is systemic progression. Patients with zoledronic acid have all been shown to be
HER2 positive disease have a high risk of brain effective.
metastasis. Trastuzumab need not be discontin- • Brain metastasis – management of brain
ued on isolated progression/relapse in the brain. metastases are dealt with in Chapter 16
In patients after trastuzumab failure, lapatinib (p. 278). Patients with HER2+ disease have a
is a treatment option, though currently not yet high risk of brain metastasis, particularly to
approved in the UK. It is an inhibitor of HER2 the posterior fossa.
and as well as EGFR (ErbB1) receptor. A rand- • Tumour related symptoms of fungation, dis-
omized study in patients who had been previ- charge and bleeding are treated with either
ously treated with trastuzumab showed a palliative mastectomy or radiotherapy.
130
BREAST CANCER 10
• Management of oncological emergencies such Special situations
as spinal cord compression and hypercalcae-
mia are discussed in Chapter 23 (p. 328). Paget’s disease
This is a pre-malignant condition of the nipple
Prognostic factors and outcome and areola occurring in the 5th or 6th decade.
Clinically there is erythema, dryness and fissuring
Lymph node metastasis is the most important of the nipple.
prognostic factor. The number of lymph nodes Microscopically it is characterized by Paget’s
involved has prognostic implications. Other cells located throughout the epidermis. More
important prognostic factors include tumour size than 95% of patients with Paget’s disease have
(affects the risk of nodal involvement, metastasis an associated ductal carcinoma. Fifty percent of
and survival), grade (affects survival), age (women patients with Paget’s disease have a lump of
aged <35 years have twice the risk of local recur- which more than 90% are invasive carcinoma.
rence and 1.6 times higher risk of distant metas- Of the other half of patients without a lump,
tasis compared with >35 years), hormone receptor 30% will have invasive carcinoma and 70% have
status (patients with ER+ tumours have better ductal carcinoma-in-situ.
survival), lymphatic invasion (presence of which Investigations include breast examination,
increases local recurrence and systemic recur- mammography and biopsy. Prognosis is de
rence) and HER2+vity (adverse prognosis, more pendent on the characteristics of associated
likely to respond to anthracyclines). malignancy.
Long-term prognosis can be estimated using Conservative surgery followed by radiother-
the Nottingham prognostic index (Figure 10.5) apy is the treatment of choice. The local recur-
or using online programmes such as adjuvant rence with surgery alone is 25–40% from a small
online (www.adjuvantonline.com). series. An EORTC study of complete excision
5-year survival of stage I breast cancer is with nipple-areolar complex with postoperative
85%, stage II 70%, stage III 50% and stage IV radiotherapy showed a local recurrence of 5.2%.
20%. Mastectomy is offered based on patient choice
and for multifocal disease. Management of
Screening and prevention regional nodes and recommendations for sys-
temic treatment are based on the features of
Evidence shows that screening using mammogra- underlying malignancy.
phy reduces death from breast cancer by a third.
In the UK screening is aimed at those with the Inflammatory breast cancer
greatest proven benefit which includes women Two percent of all breast cancers are inflamma-
aged 50–69 years. The benefit of screening tory. Clinically there is ill-defined erythema, ten-
for women aged 40–49 years and >70 years is derness, induration and eczema-like skin changes.
controversial. This is often misdiagnosed as a breast abscess.
Screening mammography is done every 2 years Microscopically it is characterized by the pres-
for the general population and annually for those ence of cancer cells in dermal lymphatics. All
with a strong family history of breast cancer. patients need staging investigations with CT of
In women having screening, the breast cancer the chest and abdomen and bone scan. Patients
prevalence is about 0.5% (i.e. about 1 in 200 without distant metastasis are treated with neo-
women who are screening will be diagnosed adjuvant chemotherapy followed by mastectomy
to have breast cancer). 10% of women recalled and axillary clearance and adjuvant radiother-
for further assessment with a ‘positive’ screening apy. In HER2+ patients, trastuzumab can be
mammogram are found to have a breast cancer. added along with neoadjuvant taxanes. Patients
Assessment and prevention of breast cancer in receive hormones if ER+.
women with family history of breast cancer is 25% of patients present with metastatic
given on p. 47. disease when the aim is palliative and the treat-
131
II SITE-SPECIFIC CANCER MANAGEMENT
ment principle is the same as metastatic breast Usual presentation is a painless mass (85%)
cancer. and other features include nipple retraction,
ulceration, discharge and pain. Investigations
Triple negative breast cancer include ultrasound, mammography and histo-
Triple negative breast cancer lacks the expression logical confirmation.
of oestrogen, progesterone and HER2 receptors. Primary treatment is total mastectomy with
Up to 15% of breast cancer falls into this cate- an axillary procedure similar to female breast
gory which is associated with a high rate of local cancer. Adjuvant radiotherapy indications are
and systemic recurrence. These tumours are similar to female breast cancer. Adjuvant
similar to BRCA-1 associated breast cancer. Cur- tamoxifen is indicated for ER+ tumours.
rently, these tumours are treated as other breast Metastatic breast cancer can be treated
cancers. Recent understanding of the role of with hormonal manipulation or chemotherapy.
BRCA1 gene pathway dysfunction in these 80% of oestrogen in men is produced peri
tumours has led to the ongoing research on the pherally and 20% production is from the testis.
role of platinum chemotherapy and poly (ADP- Hence, aromatase inhibitors may not be effec-
ribose) polymerase (PARP) inhibitors as poten- tive. Methods of hormonal manipulation include
tial therapeutic strategies. Data show that these tamoxifen, orchidectomy, anti-androgens and
tumours are sensitive to platinum-based chemo- aminoglutethimide.
therapy and hence a number of studies are under-
way to evaluate various platinum regimes. Breast cancer during pregnancy
Please see p. 292.
Bilateral breast cancer
It occurs in those with a strong family history of Recent advances
breast cancer and those who had breast cancer Several biological and target agents are under
at an early age. It can be either synchronous investigation either as single agents or in combi-
(tumours occur simultaneously or within 6 nation. A randomized study of bevacizumab (an
months of initial diagnosis) which occurs in less anti-VEGF) with paclitaxel showed a better
than 1% or metachronous (tumours diagnosed 6 response rate (37% vs. 21%, p < 0.001) and
months or more after initial diagnosis) which has progression-free survival (11.8 vs. 5.9 months,
an incidence of 1–2% per year on follow-up. p < 0.001) compared with paclitaxel alone.
These should be managed as two separate breast However, overall survival was similar in both
cancers. groups. An ongoing study (BEATRICE) is evalu-
ating the role of 1 year of bevacizumab along
Male breast cancer with standard adjuvant chemotherapy in triple
Male breast cancer accounts for 1% of all breast negative breast cancer.
cancers and 0.1% of male cancer deaths. The Other agents under evaluation include sunitinib
average age at diagnosis is 10 years later than (a tyrosine kinase inhibitor), PARP inhibitors,
women. Alterations in testosterone and oestro- cetuximab (EGFR inhibitor) and ixabepilone (a
gen balance may play a role in the aetiology microtubule inhibitor).
as illustrated by the increased risk in men
with undescended testis, or after orchiectomy. Further reading
Klinefelter’s syndrome has 50-fold increase in Veronesi U, Boyle P, Goldhirsch A, Orecchia R, Viale G.
breast cancer. Other risk factors include obesity, Breast cancer. Lancet. 2005;365:1727–1741.
cirrhosis, family history of breast cancer, Benson JR, Jatoi I, Keisch M et al. Early breast cancer.
BRCA1/2 mutations. Lancet. 2009;373:1463–1479.
Buchholz TA. Radiation therapy for early-stage breast cancer
90% tumours are invasive of which 80%
after breast-conserving surgery. N Engl J Med.
tumours are infiltrating ductal carcinoma, 5% 2009;360:63–70.
papillary and 1% lobular. 10% tumours are Pagani O, Senkus E, Wood W et al. International guidelines
DCIS. 80% tumours are ER+ and 75% PR+. for management of metastatic breast cancer: can
132
BREAST CANCER 10
metastatic breast cancer be cured? J Natl Cancer Inst. treatment and current lines of research. Cancer Treat
2010;102:456–463. Rev. 2010;36:206–215.
Roy V, Perez EA. Beyond trastuzumab: small molecule Fentiman IS, Fourquet A, Hortobagyi GN. Male breast
tyrosine kinase inhibitors in HER-2-positive breast cancer. Lancet. 2006;367:595–604.
cancer. Oncologist. 2009;14:1061–1069. Dawood S, Ueno NT, Cristofanilli M. The medical treatment
Bosch A, Eroles P, Zaragoza R, Viña JR, Lluch A. Triple- of inflammatory breast cancer. Semin Oncol. 2008;35:
negative breast cancer: molecular features, pathogenesis, 64–71.
133
Cancers of the
gastrointestinal system
TV Ajithkumar and E de Winton
11
Oesophageal cancer The common malignant tumours are adeno-
carcinoma, squamous cell carcinoma (SCC) or
Epidemiology undifferentiated carcinoma. Most adenocarcino-
Oesophageal cancer is an aggressive cancer with mas occur at or immediately above the GOJ
an overall survival rate of less than 10%. There which are classified (Siewert Classification) as
are over 7000 new patients per year in the UK follows:
and 16,000 per year in USA. The median age at • Type I – distal oesophageal cancer which may
diagnosis is 69 years. The male to female ratio is infiltrate GOJ.
2.5 : 1; but cervical oesophageal cancer is more • Type II – straddles the GOJ (cancer of the
common in women. cardia; also called junctional cancer).
• Type III – subcardial cancer which may infil-
Aetiology trate OGJ and distal oesophagus.
Oesophageal reflux and Barrett’s oesophagus are
Adenocarcinoma – accounts for 65% of
thought to increase the risk of adenocarcinoma.
oesophageal cancers in the UK and the major
Oesophageal reflux may account for the increased
aetiological factor is Barrett’s oesophagus. The
incidence of adenocarcinoma (10% per year) in
incidence of carcinoma arising in Barrett’s
the Western world.
oesophagus is 1 per 100 patient-years.
Box 11.1 shows risk factors for oesophageal
Squamous cell carcinoma (SCC) accounts for
cancer. A diet rich in fruit and vegetables has
25% of oesophageal cancer. Alcohol and smoking
been shown to reduce the relative risk.
are the major etiological factors. SCC is common
in upper and mid third cancers.
Anatomy
Most common sites of metastatic spread are
The oesophagus extends from the cricopharyn- lymph nodes (70%), lung (20%), liver (35%),
geal sphincter to the gastro-oesophageal junction bone (9%) and adrenal glands (2%).
(GOJ) and is 25 cm in length. Figure 11.1 shows
the anatomic sections of the oesophagus. The
majority of tumours (85%) arise in the middle Clinical features
and lower third of oesophagus and 15% arise in Most patients present with progressive dysphagia
the upper third. and difficulty in swallowing liquids indicate an
advanced stage. 50% patients experience painful
Pathogenesis and pathology swallowing and other presentations include
Oesophageal cancer is thought to be the end reflux, regurgitation and vomiting following
result of a multi-step process of carcinogenesis. eating, rapid weight loss. Local invasion of cancer
Adenocarcinoma arising from Barrett’s oesopha- leads to pain, and nerve compression leading to
gus is thought to undergo a stepwise progression Horner’s syndrome, recurrent laryngeal nerve
with associated genetic changes. palsy and a raised hemi-diaphragm.
134 © 2011, Elsevier Ltd
DOI: 10.1016/B978-0-7234-3458-0.00016-6
CANCERS OF THE GASTROINTESTINAL SYSTEM 11
Cervical
15–18 cm from incisors 15% of cancers
•
Squamous cell carcinoma
Upper third thoracic
common
18–24 cm from incisors
Figure 11.1
Anatomic section of oesophagus.
135
II SITE-SPECIFIC CANCER MANAGEMENT
Radical surgery
• Other investigations – include full blood Surgery is the best single modality treatment
count, biochemistry and coagulation profile. compared with radiotherapy alone. The best
Patients considered for surgery need pulmo- outcome from specialist centres is 5-year survival
nary function tests, arterial blood gases, ECG of approximately 35% with a postoperative mor-
and an exercise test. tality of less than 5%.
Type of surgery depends on the site of
Staging tumour, histology and the extent of lymphade
Staging determines prognosis and guides treat- nectomy required. The surgery involves subtotal
ment. TNM staging is given in Box 11.2. oesophagectomy with the upper part of the
stomach and anastomosis of the cervical oesopha-
Management of oesophageal cancer gus and gastroplasty. 60–70% of patients have
(Figure 11.3) involved lymph nodes at the time of surgery and
Localized disease hence need dissection of abdominal and medias-
tinal nodes in middle and lower third cancers (a
This group consists of patients with stages I–III
minimum of 15 nodes should be removed).
oesophageal cancer as well as a subgroup of
The common procedures are as follows:
operable stage IVa (involving pleura, pericar-
dium and diaphragm). However, only one-third • Upper third oesophageal cancer: three-phase
of patients present with localized disease. The approach with complete removal of the
majority of these are stage II or III disease and tumour and mediastinal and supraclavicular
less than half of these patients are curable. Assess- lymphadenectomy.
137
II SITE-SPECIFIC CANCER MANAGEMENT
• Barium study
• Endoscopy and biopsy
• EUS
• CT scan chest/abdomen/pelvis
• Blood counts and biochemistry
Localized Metastatic
• Radical chemoradiotherapy Fit for surgery Unfit for surgery or unresectable Unfit for surgery and
• Radical radiotherapy and fit for chemoradiotherapy chemoradiotherapy
• Palliative
• Bronchoscopy if indicated
• Laparoscopy for distal third tumours
• PET scan
Figure 11.3
Management of oesophageal cancer.
• Middle and lower third cancers: two stage apy is contraindicated. The best results are an
Ivor–Lewis approach. The first stage consists overall 5-year survival of less than 10%.
of laparotomy and mobilization of the
Radical chemoradiotherapy (CRT)
stomach, followed by right thoracotomy to
Radiotherapy is given in combination with chem-
remove the tumour and to form an oesoph-
otherapy which is proven to be superior to radical
agogastric anastomosis.
radiotherapy alone. Chemotherapy improves
Minimally invasive surgical approaches are local control by radiation sensitization and helps
evolving. to minimize distant metastatic relapses. This
approach results in an overall 3-year survival of
Radical radiotherapy (Box 11.3) 30%. The commonly used combination is cispla-
Radical radiotherapy is the treatment of choice tin with 5-fluorouracil with radiotherapy at a
in patients with localized disease who are medi- dose of 50 Gy in 2 Gy per fraction (Box 11.3).
cally unfit for surgery and in whom chemother- However, locoregional recurrence remains the
138
CANCERS OF THE GASTROINTESTINAL SYSTEM 11
major cause for overall failure with 50% of Preoperative chemotherapy followed
patients developing locoregional disease. by surgery
Attempts to increase the radiotherapy dose to A meta-analysis showed that preoperative chem-
improve local control has resulted in increased otherapy followed by surgery improved 2-year
treatment-related deaths without improvement survival by 7% compared with surgery alone
in local control or survival. Studies comparing (HR 0.90; CI 0.81–1.00). The largest study
CRT with CRT followed by surgery showed (MRC OE02) of 802 patients which compared
similar overall survival, but with high treatment- surgery alone with two cycles of 3-weekly preop-
related mortality with surgery. erative cisplatin (80 mg/m2 on day 1 as 4 hour
139
II SITE-SPECIFIC CANCER MANAGEMENT
infusion) and 5-fluorouracil (1 g/m2/day continu- study (REAL-2) to explore the role of oxaliplatin
ous infusion for 4 days) followed by surgery instead of cisplatin and capecitabine instead of
showed that chemotherapy improves overall sur- 5-FU (EOX regime) showed that toxicity of 5FU
vival from 34% to 43% at 2-years (p = 0.004). and capecitabine were similar; oxaliplatin resulted
Hence, this is the standard treatment option in in increased neuropathy and diarrhoea but
most UK centres. reduced renal toxicity, thromboembolism, alo-
pecia and neutropenia when compared with cis-
Preoperative CRT followed by surgery platin. Patients receiving EOX survived longer
A meta-analysis showed that preoperative CRT than ECF (38% vs. 47% at 1-year; p = 0.02).
followed by surgery improved 2-year survival
by 13% compared with surgery alone (HR 0.81 Palliative treatment of dysphagia
95% CI 0.70–0.93; p = 0.002). However, this Dysphagia is the predominant symptom which
benefit was only seen with concurrent chemora- needs palliation. The various methods to relieve
diotherapy rather than sequential and treatment- dysphagia are:
related mortality was increased with preoperative
• Endoscopic dilatation – results in immediate
CRT (p = 0.053). Due to concerns about mor-
but short lived (2–4 weeks) improvement and
bidity and mortality, it is not practised in the UK.
Figure 11.4
Recurrence
Management of recurrence.
Locoregional Metastatic
• Endoscopic resection Medically fit for Medically fit but Medically unfit for
• Gastric resection and resection unresectable resection
lymphadenectomy
Laparoscopy
3 courses of
chemotherapy with ECF Yes No
Surgery
Recurrence or progression
Perioperative treatment
Box 11.6: Treatment regimes in gastric cancer
A randomized study (UK MRC MAGIC) showed
Perioperative chemotherapy and surgery
that three cycles of pre- and postoperative chem- (UK MRC MAGIC trial)
otherapy with epirubicin, cisplatin and continu- • ECF chemotherapy: Epirubucin 50 mg/m2 IV day 1,
ous infusion 5 fluorouracil (ECF) significantly cisplatin 60 mg/m2 IV day 1 and 5-fluorouracil
200 mg/m2/day continuous IV infusion for 21
improved 5-year survival (36% vs. 23%; days. Cycle repeated 3-weekly. Total 3 cycles
p = 0.009) compared with surgery alone in preoperatively.
resectable gastric and lower oesophageal cancers • Surgery 3–6 weeks after chemotherapy
(Box 11.6). This is the standard of care in most • Postoperative chemotherapy – 3 cycles of ECF
of the UK and parts of the Europe (Box 11.6). started 6–12 weeks after surgery
Metastatic or unresectable cancer
Unresectable gastric cancer ECF regime (see above)
The median survival of patients with unresecta- ECX regime
• Epirubicin 50 mg/m2 day 1, cisplatin 60 mg/m2/day
ble non-metastatic cancer is 6 months without 1 and capecitabine 625 mg/m2 orally twice daily;
treatment. Treatment is aimed at improving cycle repeated 3-weekly
the symptoms and quality of life and possible
extension of life. The various palliative measures
include:
outlet obstruction. Endoscopic stent place-
• Bleeding – endoscopic laser photocoagulation ment offers effective palliative. However,
or argon plasma coagulation is effective in 15–40% with develop recurrent symptoms.
controlling bleeding. If these measures fail, An alternative option is palliative bypass.
palliative resection may be considered. • Radiotherapy – is effective in controlling
• Obstruction – tumours of the proximal bleeding or obstruction in patients who are
stomach may present with dysphagia whereas unsuitable candidates for other interventions.
distal stomach tumours present with gastric Radiotherapy is effective in controlling pain.
146
CANCERS OF THE GASTROINTESTINAL SYSTEM 11
Systemic treatment of unresectable Screening
gastric cancer Routine screening is not recommended for gastric
Patients with unresectable and/or disseminated cancer. Role of screening of asymptomatic indi-
gastric cancer should be considered for a clinical viduals for gastric cancer from countries with
trial, and those with good performance status high incidence such as Japan shows inconsistent
(0–2) may be offered systemic therapy. Rand- results of benefit.
omized trials suggest that chemotherapy may
improve survival compared with best supportive Gastric stump cancer
care (9–11 months vs. 3–4 months HR 0.39) and By definition this is the cancer developing in the
quality of life. A meta-analysis showed that gastric remnant at least 5 years after surgery for
combination chemotherapy improves survival benign peptic ulcer disease. Patients after distal
by 6 months and the best survivals are achieved gastric resection have a 4–7 fold increase in
by three-drug regimens containing 5-FU, anthra- gastric cancer attributed mainly to gastroduode-
cycline and cisplatin. The standard regime used nal reflux. Treatment is complete removal of
in the UK is ECF (Box 11.6). A recent study gastric remnants with a D2 lymphadenectomy.
(REAL 2) showed that ECF is comparable to The pattern of lymph node metastases in these
EOX. This regimen has the advantage of avoiding cancers may differ from primary gastric cancer
the need for continuous infusion of 5-FU. and the prognosis is the same as primary gastric
Other active agents include taxanes, and cancer.
irinotecan.
Follow-up Cancers of the liver
There is no evidence that regular intensive fol- Introduction
low-up improves outcome and hence symptom
driven follow-up is recommended. Cancers of the liver can be primary (5%) or sec-
ondary (95%). The most common primary liver
Management of recurrence cancer is hepatocellular carcinoma (HCC) which
The role of second-line chemotherapy is not well- accounts for 90% of primary liver cancers. There
established. The commonly used agents include are more than 3000 new patients diagnosed in
taxanes, irinotecan and oxalipatin. In the major- the UK. Males are more commonly affected
ity of patients management is essentially pallia- (male: female ratio is 5 : 3). Median age of diag-
tive aiming at best supportive care. nosis is 64 years.
Prognosis Aetiology
Important prognostic factors in resectable gastric The risk factors include:
cancer include depth of invasion, number of • Chronic liver disease – 80–90% HCC arising
lymph nodes involved and positive resection in patients with cirrhosis.
margins. Estimated 5-year survival by stage is: • Hepatitis infection – hepatitis B and C viruses
• Stage I: 70%. account for 75% cases of HCC. HCV accounts
• Stage II: 40%. for the recent rising trend.
• Stage III: 20%. • Alcohol accounts for 10% of cancers in Asia
• Stage IV: less than 5%. and 20% in Europe and USA.
• Non-alcoholic fatty liver disease (NAFLD) –
Newer agents associated with obesity and type 2 diabetes.
Monoclonal antibodies and various agents tar- 5% patients progress to cirrhosis and have an
geting pathways of cellular proliferation are increased risk of HCC.
being evaluated. However data are still limited • Aflatoxin.
to early phase studies. Examples of molecules • Metabolic disorders – haemochromatosis,
under study are bevacizumab (anti-VEGF), cetix- alpha-1-antitrypsin deficiency, and Wilson’s
umab (anti-EGFR) etc. disease.
147
II SITE-SPECIFIC CANCER MANAGEMENT
HCC
Very early stage (O) Early stage (A) Intermediate stage (O) Advanced stage (C) Terminal
1 HCC <2 cm 1 HCC or 3 nodules Multinodular Portal invasion stage (D)
Carcinoma in situ <3 cm, PST 0 PST 0 N1, M1, PST 1–2
1 HCC 3 nodules ≤3 cm
Portal pressure/bilirubin
Increased
No Yes No Yes
Figure 11.9
Staging classification and treatment of hepatocellular carcinoma (Llovet JM, Burroughs A, Bruix J. LANCET
2003;362;1907–1917, fig 5, with permission). PST – performance status test, PEI – percutaneous ethanol injection.
Liver transplantation
Box 11.8: Child–Pugh grading of severity of
liver disease Liver transplantation is a treatment option for
HCC in cirrhotic patients. Indications for liver
Score
transplantation (only 10% are eligible) include
Criteria 1 2 3
(Milan criteria):
Hepatic None 1–2 3–4
encephalopathy • Solitary HCC <5 cm.
(grade) • Up to three HCC all <3 cm.
Total bilirubin (mg/dl) <2 2–3 >3
Ascites Absent Mild Moderate In these highly selected patients the reported
or severe 5-year survival is >70%. Liver transplantation
Serum albumin (g/dl) >3.5 2.8–3.5 <2.8
is not an acceptable option for HCC in non-
Prothrombin time <4 4–6 >6
(second prolonged) or or or cirrhotic patients because of the need for intense
or INR 1.7 1.7–2.3 >2.3 immunosuppression.
Child–Pugh Grade A = 5–6 points.
Grade B = 7–9 points. Percutaneous ablation
Grade C = 10–15 points. Percutaneous ablation is a treatment option for
patients with stage 0 & A disease who are not
149
II SITE-SPECIFIC CANCER MANAGEMENT
suitable for resection or transplantation and • Stage 0 & A after radical treatment – 5-year
those who are waiting for transplantation. Abla- survival 50–75%.
tion is achieved either by: • Stage B – median survival 16 months without
treatment and with TACE 3-year survival is
• Chemical injection (ethanol, acetic acid and
50%.
boiling saline).
• Stage C – median survival 6 months without
• Thermal ablation (radiofrequency, laser and
treatment and with palliative treatment <10%
cryotherapy).
survive for 3 years.
Percutaneous ethanol injection (PEI) and radi- • Stage D – 1 year survival <10%.
ofrequency ablation (RFA) are equally effective
for tumours of <2 cm whereas RAF is more effec- Screening
tive for larger tumours. Surveillance is offered to patients with cirrhosis
and the two most common tests used for
Stage B & C screening are serum alpha-foetoprotein (AFP)
Treatment is palliative and options include: and ultrasonography.
Figure 11.10
Extent of dissection in radical
cholecystectomy.
Gall bladder
Nodes of hiatus
Nodes along
hepatic artery
Cystic lymph node
Pericholedochal
lymph nodes
Treatment
• Types I and II: en bloc resection of the extrahepatic bile ducts and gall bladder, regional
lymphadenectomy, and Roux-en-Y hepaticojejunostomy
• Type III: as above plus right or left hepatectomy
• Type IV: as above plus extended right or left hepatectomy
• Distal cholangiocarcinomas are managed by pancreatoduodenectomy as with ampullary or
pancreatic head cancers
• The intrahepatic variant of cholangiocarcinoma is treated by resection of the involved
segments or lobe
153
II SITE-SPECIFIC CANCER MANAGEMENT
A B
Figure 11.12
CT scan and ERCP in pancreatic cancer. Carcinoma pancreas (white arrow) leading biliary obstruction (black arrow)
and duodenal obstruction with gastric dilatation (white asterix) (A) and ERCP shows strictures of common bile duct
and pancreatic duct (‘double duct sign’) (B).
155
II SITE-SPECIFIC CANCER MANAGEMENT
and specificity of 90%. It is useful to assess Patients with jaundice may be considered for pre-
response and identify tumour recurrence. A operative biliary drainage. The type of surgery
level of <200 u/ml suggests longer survival. depends on location of the tumour:
Further tests prior to surgery • Tumours of head of the pancreas – pan
creaticoduodenectomy (classic Whipple) or
Laparoscopy with laparoscopic ultrasound alters
pylorus preserving pancreatoduodenectomy
the management of 15% of patients already
(pp Whipple) are considered curative. A meta-
assessed as resectable by CT. Selective laparos-
analysis showed that both procedures carry
copy based on the serum level of CA19-9 and
equal morbidities and survival. There is no
large tumour, is a more efficient strategy, reduc-
survival advantage for extended radical
ing the proportion of patients undergoing laparo-
lymphadenectomy in pancreatic cancer.
scopic ultrasound from 100% to around 45%
• Tumours of the body and tail of pancreas –
while increasing the yield from 15% to 25%.
left pancreatectomy which includes splenec-
Staging tomy and en bloc removal of the hilar lymph
Staging is shown in Box 11.10. nodes.
The postoperative morbidity of radical surgery
Management of pancreatic cancer
is around 40% and the mortality is <5%.
Treatment of resectable cancer
Neoadjuvant therapy
Surgery
Neoadjuvant treatment using a combination
Patients with good performance status and
of external beam radiotherapy with 5-FU or
resectable tumours are treated with radical resec-
Gemcitabine based chemotherapy may improve
tion. Criteria for resectability are given in Box
resectability in a borderline resectable tumour.
11.11. Though the aim of surgery is a complete
Several studies report a resection rate of 60%
microscopic resection (R0), 30–60% of resec-
and negative resection margin of 90%. However,
tions result in microscopic residual disease (R1).
this is not routinely practised in the UK.
Postoperative treatment
• Adjuvant chemotherapy – a meta-analysis
showed that adjuvant chemotherapy improves
Box 11.10: Staging of pancreatic cancer 5 year survival (19% vs. 12%) and median
Stage 0 survival (19 months vs. 13.5 months) com-
TisN0M0 Carcinoma-in-situ pared with no chemotherapy. The recently
Stage IA
reported ESPAC-3 study comparing gemcit-
T1N0M0 Tumour limited to the pancreas, ≤2 cm in abine and 5-FU/FA in patients who had R0/
diameter
Stage IB
T2N0M0 Tumour limited to the pancreas, >2 cm in
diameter
Stage IIA Box 11.11: Criteria for resectable
T3N0M0 Tumour beyond pancreas, but without pancreatic cancer
involvement of coeliac axis or superior mesenteric
artery • No coeliac, hepatic or superior mesenteric artery
involvement
Stage IIB • A patent superior mesenteric-portal venous
T1–3N1M0 Regional lymph node metastasis confluence
Stage III • Portal venous involvement of not more than 2 cm
T4 any NM0 Tumour involving coeliac axis or in length or more than 50% circumference
superior mesenteric artery • No liver, peritoneal or other distant metastases
Stage IV • Absence of portal hypertension and cirrhosis
Any T any N M1 Distant metastasis • No severe co-morbidity to exclude surgery
156
CANCERS OF THE GASTROINTESTINAL SYSTEM 11
R1 resection showed no difference in survival Symptom control
(median survival of 23.0 months with 5-FU/ • Pain control – measures include analgesics,
FA and 23.6 months with gemcitabine). celiac plexus block or unilateral thoraco-
• Adjuvant chemoradiotherapy – a meta-analy- scopic splanchnicectomy.
sis showed that chemo-radiotherapy did not • Obstructive jaundice – managed with biliary
improve survival compared with no treatment stent or surgical bypass. Plastic stents are used
(median survival 15.8 months vs. 15.2 for patients with metastatic disease and
months). tumours of >3 cm diameter whereas self-
• Adjuvant chemoradiotherapy and chemo expanding metal stents are used for patients
therapy – RTOG 9704 trial reported no with good performance status and locally
statistically significant survival benefit advanced disease of <3 cm.
with gemcitabine to adjuvant 5-FU based • Duodenal obstruction – treated with endo-
chemoradiotherapy. scopically placed stents or bypass surgery.
• Gastrinoma presents with refractory multiple The clinical presentation of a carcinoid tumour
peptic ulcers and diarrhoea. It is diagnosed by can be a non-specific abdominal symptom,
high gastrin levels (>100 pg/ml, or >200 pg/ although carcinoid syndrome can occur in 25%
ml after secretin stimulation). of cases. CT is the diagnostic image of choice.
• Glucagonoma presents with features of hyper- Surgery is the treatment of choice for resectable
glycaemia, stomatitis, weight loss, diarrhoea tumours. However, 70–80% patients present
and psychiatric disturbances. Necrolytic with advanced disease. Treatment options for
migratory erythema of the skin is a character- advanced disease include:
istic feature. Diagnosis is by high plasma glu-
• Symptom control using octreotide or a long
cagon levels.
acting version.
• Vasoactive intestinal polypeptide tumour
• Radioisotope treatment using 131I-MIBG or
(VIPoma) causes watery diarrhoea and 111
In/90Y octreotide in patients with positive
hypokalaemia. Diagnosis is by high plasma
scans.
levels of vasoactive intestinal polypeptide.
• Palliative chemotherapy – streptozosin plus
Staging investigations of pancreatic endocrine 5-FU/dacarbazine and doxorubicin plus 5-FU.
tumours include CT scan, MRI scan, EUS, 111In-
The overall five-year survival rate is 30–40%
octreotide and selective portal/splenic venous
and the median survival of patients with meta-
sampling and intraoperative ultrasound.
static disease is approximately 7 months.
Surgical resection is the treatment of choice
which may be indicated even in metastatic
disease. 5-year survival following surgical resec- Malignant tumours of
tion is 50–95%. the small intestine
Cystic tumours of pancreas Small intestinal malignancies constitute 2–3% of
all malignant GI cancers. The ileum is the most
These constitute around 15% of all pancreatic
frequent site of tumours and is more common
cystic masses. The common types are:
in males.
• Serous cystic neoplasms predominantly affect
women, are found mostly in the head of the Aetiology
pancreas and represent 30% of cystic neo- The risk factors include:
plasms. Conservative approach with regular
• Familial adenomatous polyposis (FAP) – small
imaging is the management.
intestinal adenocarcinoma is one of the
• Mucinous cystic neoplasms are also common
common causes of death in patients with FAP
in women affecting the body and tail of the
after colectomy.
pancreas, and represent 40% of primary
• Crohn’s disease (adenocarcinoma).
cystic neoplasms. These are treated with
• Coeliac disease (adenocarcinoma and lym-
resection to avoid malignant transformation.
phoma) – type of lymphoma in coeliac disease
• Intraductal papillary mucinous neoplasms
is enteropathy associated T cell variant.
(IPMNs) commonly affect men and constitute
• Tropical sprue (lymphoma).
30% of pancreatic cysts. IPMNs arising from
• MEN type-1 (gastrin producing tumour of
main duct should be resected whereas those
duodenum and jejunum).
from branch duct may be managed with
regular follow-up imaging. Pathology
Carcinoid tumours Tumours of small intestine can be of epithelial,
mesenchymal or lymphoid origin. The following
Carcinoid tumours are neuroendocrine neo-
are the common types of cancers occurring in the
plasms arising from enterochromaffin cells. These
small intestine:
can be functioning or non-functioning. The mean
age at presentation is 49 years and women are • Adenocarcinomas (45%) – occur in ampulla
more commonly affected. of Vater in the duodenum and jejunum.
158
CANCERS OF THE GASTROINTESTINAL SYSTEM 11
• Carcinoids or neuroendocrine tumours (30%) Staging
– common in terminal ileum. Adenocarcinoma is staged similar to colonic
• Sarcomas (10%) – common in ileum, major- cancer (p. 163). Lymphomas are staged using the
ity of tumours are GIST (p. 261). Ann Arbor staging (p. 299).
• Lymphomas (15%) – B cell NHL is the most
common. Management
• Metastatic cancers – are commoner than Small intestinal adenocarcinoma
primary and usual primaries are GI tract,
In patients with localized adenocarcinoma, resec-
breast, uterus, ovary and melanoma.
tion of visible disease and regional lymph nodes
Clinical features is done. In advanced disease, surgery may help
Clinical presentation depends on the type of with palliation of symptoms.
cancer, site and size. The majority of patients Chemotherapy regime is same as that of
with small intestinal malignancies present with colonic cancer (p. 165). A small study reports a
advanced disease. Nonspecific symptoms include response rate of 50% and a median survival of
abdominal pain, anaemia, nausea, bleeding, and 20 months with a combination of oxaliplatin
weight loss. Patients can also present as a surgical with capecitabine.
emergency with perforation or intussusception. Small intestinal lymphoma
Duodenal tumours can present with obstruction
as well as jaundice. Metastatic liver carcinoid can Stage I and II B-cell NHL are treated surgically
present with carcinoid syndrome. Lymphoma with or without chemotherapy, whereas III–IV
may present with pain, weight loss and features disease is treated with primary chemotherapy
of malabsorption. GIST commonly presents with with or without surgical debulking. Chemother-
anaemia, sometimes with an abdominal mass. apy regimens are similar to NHL (p. 305).
Investigations GIST
The initial investigations depend on the present- Managed similar to GIST occurring elsewhere
ing symptoms. (p. 261).
Imaging Prognosis
• Plain X-rays – useful in suspected Prognosis depends on the type of cancer and
obstruction. stage. Adenocarcinoma usually presents with
• Abdominal ultrasound – useful to detect liver advanced disease and the reported 5-year sur-
metastasis, ascites and biliary dilatation. vival is 30%. Duodenal tumours have a better
• CT scan of chest, abdomen and pelvis can prognosis with 5-year survival of 50%.
delineate primary tumour and disease extent. Lymphomas have a 10-year survival of 60%.
Different histological types exhibit different The prognosis of small bowel GIST depends on
features, e.g. lymphoma appears as diffuse their size and mitotic rate (see p. 262 for NIH
segmental thickening of the small intestine prognostic index).
and GIST as a well circumscribed mass.
• Small bowel follow-through. Colorectal cancer
• Endoscopic ultrasound (EUS).
Epidemiology
• Small bowel endoscopy.
• 111In-octreotide scan – in carcinoid to rule out Colorectal cancer (CRC) is the third commonest
metastatic disease. cancer in the UK after breast and lung. Over
36,000 new cases per year are diagnosed in the
Tissue biopsy UK and 105,500 per year in the USA.
Biopsy confirmation is essential prior to defini- Over 80% of cases occur in people over 60
tive treatment. Tissue diagnosis can be obtained and is rare below the age of 40 (except in heredi-
by endoscopic, CT guided or laparoscopic tary forms). The male:female incidence ratio is
methods or by laparotomy. 1.2 : 1.0. In the UK, the lifetime risk is 1 in 18
159
II SITE-SPECIFIC CANCER MANAGEMENT
for men and 1 in 20 for women. Two-thirds of • Drugs – females on hormone replacement
primaries are in the colon and one third in the therapy and those using regular aspirin are at
rectum. Left-sided cancers are more common low risk of developing CRC.
than right (sigmoid and rectal cancers account • Lifestyle – long-term smokers are at high risk
for over half). of adenomas and CRC mortality. Regular
physical activity reduces the risk of CRC, par-
Aetiology ticularly in men.
The majority of colon cancers are sporadic and
not associated with known hereditary genetic
mutations. Hereditary bowel cancer is character-
Pathogenesis and pathology
ized by early age at diagnosis, right-sided cancers, Almost all colorectal cancers are adenocarcino-
synchronous/metachronous colorectal tumours mas and arise in adenomatous polyps through a
or other characteristic tumours in same individu- multi-step process (Figure 11.13). Most adeno-
als or families (see below). Up to 25% of patients carcinomas are moderate to well differentiated
with CRC give a positive family history, suggest- with typical morphology. About 80% of primary
ing involvement of genetic factors. colorectal adenocarcinomas and 70% of
metastases will be CK7− CK20+. Rare histologic
• Genetic (p. 50): types include squamous cell carcinoma, small
• Familial adenomatous polyposis (FAP) cell carcinoma, adenosquamous carcinoma and
accounts for 1% of all cases of CRC. medullary carcinoma.
• Hereditary non polyposis colorectal cancer
(HNPCC) accounts for about 5% of all
CRC cases. Presentation
• Familial – those with an affected first-degree About 85% of patients with bowel cancer have
relative are at increased risk of developing these symptoms at the time of diagnosis. Approxi-
CRC (relative risk 2.25). mately one-third presents with altered bowel
• Inflammatory bowel disease – increases the habit or obstructive symptoms (usually left sided
risk. In ulcerative colitis the cumulative risk tumours as the distal bowel is narrower and faecal
of developing CRC is estimated to be 8% at content more solid) and one-third with iron defi-
20 years and 18% at 30 years. ciency anaemia (usually right-sided tumours).
• Diet and supplements – high intake of red For rectal cancers other symptoms include faecal
meat, calorie and high body mass index incontinence, passage of mucus and tenesmus. Up
increase the risk whereas high vegetable intake to 30% present with acute colonic obstruction.
and high-fibre diet are protective. More advanced tumours may cause weight loss,
Figure 11.13
Common genetic mutations Consequences
Pathogenesis of colorectal cancer.
A B
Figure 11.14
Double contrast barium enema shows a
polypoid cancer with an irregular indrawn
base (A), colonoscopy showing T1 high rectal
cancer (B) and CT colonography showing an
annular carcinoma (C). Parts A and C from
Adam A, et al: Grainger & Allison’s
C Diagnostic radiology, 5th Edition, Volume 1,
2009 (Elsevier), with permission.
161
II SITE-SPECIFIC CANCER MANAGEMENT
Figure 11.15
• Surgery.
T2W MRI of mid rectal cancer shows threatened • Consider adjuvant chemotherapy in all
mesorectal margin. patients with PS 0–2.
A B
Figure 11.16
CT scan of liver in portal venous phase shows resectable liver metastases (A) and MRI liver with hepatocyte specific
contrast reveals more extensive metastases (B).
162
CANCERS OF THE GASTROINTESTINAL SYSTEM 11
Mucosa • Palliative chemotherapy
• Palliative radiotherapy
• Other palliative treatments
• Active symptom control
Lamina propria Surgery
Muscularis mucosa Surgery is curative and several studies have
shown equal survival with better cosmesis and
Submucosa shorter hospital stays for laparoscopic versus
open surgery for colectomies. Emergency surgery
has higher perioperative mortality than that of
elective surgery. Hence emergency stenting may
Muscularis propria be useful to temporarily decompress an obstructed
lesion prior to definitive surgery.
Adjuvant chemotherapy
Subserosa Table 11.1 shows current recommendations for
Serosa adjuvant chemotherapy. There is no benefit of
adjuvant chemotherapy in Dukes’ A disease. In
Mesorectal tissue/mesorectal stage III (Dukes’ C) disease, 5-fluorouracil/folinic
membrane in rectum acid (5FU/FA) chemotherapy gives an absolute
Tis – confined to mucosa or lamina propria survival benefit of between 8–13%, with 6 months
(no significant metastatic potential) chemotherapy as effective as 1 year. In stage II
T1 – invades submucosa disease, chemotherapy results in a similar propor-
T2 – invades muscularis propria
T3 – invades subserosa/pericolic/mesorectal tissue tional reduction (approximately 33%) in recur-
T4 – invades other organs or tumour on serosal surface rence, but absolute benefits are less because of
lower mortality rates (QUASAR 1 showed an
Stage group TNM stage Dukes stage % Patients absolute increase in 5-year OS of 3.6% for patients
O Tis N0 M0 – less than 70 years). Patients with stage II disease
with adverse features have a higher disease-related
I T1-2 N0 M0 A 11
mortality rate (with >1 risk factor this approaches
IIA T3 N0 M0 that of Dukes’ C cancers) and hence absolute
B 34
IIB T4 N0 M0 benefits of adjuvant chemotherapy are greater.
Capecitabine has been shown to have equal
IIIA T1-2 N1 M0
efficiency with favourable toxicity profiles com-
IIIB T3-4 N1 M0 C1 or 2 26 pared to bolus 5FU in patients with stage III
IIIC T1-4 N2 M0 disease. This is now standard of care for high
IV T1-4 N1-2 M1 D 29
risk Dukes’ B cancers and those patients with
Dukes’ C not fit for combination chemotherapy.
In patients with stage II and III CRC, addition of
N1 = 1–3 lymph nodes (LN) contain metastatic carcinoma,
N2 = ≥4 LN contain metastatic carcinoma, oxaliplatin to 5FU/FA improved 3-year DFS by
C1 = apical LN negative, 5% over infusional 5FU/FA alone at the cost of
C2 = apical LN positive increased neurotoxicity. Absolute benefit was
Figure 11.17 greater for stage III vs. stage II disease (6.9% vs.
Staging for colorectal cancer. 2.7%). Hence oxaliplatin in combination with
5FU/FA is a treatment option for stage III colon
Stage IV cancer.
• Consider radical surgery with perioperative Ideally chemotherapy should begin within six
chemotherapy if resectable liver +/− lung weeks of surgery dependent on wound healing
metastases. and patient recovery. The benefit of adjuvant
163
II SITE-SPECIFIC CANCER MANAGEMENT
Consider stenting or
palliative resection/by-
pass surgery as
appropriate
(de-functioning stoma
rarely required)
Surgical No Yes
follow-up
alone
Adjuvant Peri-operative Palliative • Best supportive care
chemotherapy chemotherapy chemotherapy • Other palliative treatments
* <4 lung resectable metastases following cardiothoracic surgical review and/or liver metastases resectable or potentially resectable with downstaging Ox/MdG
chemotherapy following liver surgeons review (requires >30% of liver intact post surgery. Note that age, number, size and proximity of metastases to major vessels
are not exclusions to this) Patients WHO PS 0-1
Figure 11.18
Management of colon cancer.
treatment starting beyond 3 months of surgery is their primary disease (Figure 11.16). However,
uncertain. resection is feasible only in 10% of patients with
metastatic disease. Preoperative chemotherapy,
Management of isolated lung or in selected patients whose liver metastases are
liver metastases initially extensive for surgery, can enable curative
Liver or lung metastasectomy should be consid- resection in about 30%. A maximum of 12 weeks
ered in fit patients following radical resection of treatment is recommended prior to surgery as
164
CANCERS OF THE GASTROINTESTINAL SYSTEM 11
Figure 11.19
Adenocarcinoma of rectum without evidence
Management of rectal cancer.
of distant metastasis and fit for surgery
Yes No
Radical intent
Consider neoadjuvant
MRI predicts R0 chemotherapy as first Manage as for colon
Radical intent
resection treatment as for colon cancer (Fig 11.18)
cancer
* In patients not receiving concurrent neoadjuvant fluropyrimidine chemotherapy manage as for colon cancer according to Dukes stage
by PET before committing patients to such useful to control bleeding in colonic and previ-
morbid surgery. If preoperative CRT is previ- ously irradiated rectal cancers.
ously not given, it should be used to decrease the
risk of further recurrence. Re-irradiation is not a Radiotherapy
standard practice but may be considered in For unresectable rectal cancer radiotherapy can
selected cases. offer symptom control, particularly for bleeding,
tenesmus and pain.
Palliation of colorectal cancer Palliative chemotherapy
Unresectable locally advanced or recurrent Chemotherapy given early, rather than after
disease and stage IV disease (excluding limited development of severe symptoms, is associated
liver and or lung metastases) is treated with with better outcomes without adverse effects on
palliative intent. quality of life.
Fluropyrimidines improve median survival by
Endoluminal procedures approximately 4 months over best supportive
Expanding metal stent can be placed endoscopi- care alone (10 vs. 6 months). The infusional
cally in upper rectal and left or right-sided colonic regimen Modified de Gramont (MdG) and oral
lesions (technically difficult to place through capecitabine have a response rate of approxi-
tumours involving the flexures). It relieves mately 30% and favourable toxicity profiles in
obstructive symptoms in 90% of patients for comparison to other 5FU/FA regimes. The addi-
more than a year (10% migration rate is 10% tion of either irinotecan or oxaliplatin to MdG
and 10% re-obstruction rate) and avoids the or capecitabine as first line treatment increases
need for a stoma. Endoscopic laser ablation is response rates to 50–60% and median survival
168
CANCERS OF THE GASTROINTESTINAL SYSTEM 11
to 14–16 months at the cost of increased toxici- treatment are to detect recurrent disease at a
ties. The addition of second line treatment pro- potentially curable stage and surveillance for
longs median survival to more than 20 months. metachronous polyps or bowel cancers. Optimum
follow up strategy is uncertain and varies widely.
Targeted systemic therapy
Routine surveillance of the bowel
Bevacuzimab (anti-VEGF) as first line in com
Patients who do not have their entire colon
bination with irinotecan/5FU increased median
imaged at diagnosis should have colonoscopy
survival by 4.7 months (20.3 m vs. 15.6 m), and
within 6 months of treatment to exclude syn-
with oxaliplatin/5FU increased 18 month survival
chronous polyps or cancers. For patients who
by 10% (63% vs. 53%), compared to respective
have had complete bowel imaging at staging,
chemotherapy alone. The common side effects of
initial colonoscopy 1–3 years following treat-
bevacuzimab include hypertension, proteinuria,
ment is recommended, followed by 5-yearly
gastrointestinal perforation, intra-abdominal
(more frequently in patients with ≥5 polyps)
infections, impaired wound healing, and increased
colonoscopy in polyp free patients with a life
arterial thromboembolic events. Treatment should
expectancy of ≥15 years.
not be given within 28 days of major surgery.
Cetuximab and panitumumab are anti-EGFR Routine surveillance for distant disease
antibodies. Cetuximab in combination with Studies suggest that intensive follow-up is associ-
irinotecan/5FU (CRYSTAL study) increased ated with significantly improved 5-year OS (HR
response rate (RR) (39% vs. 47%) and improved 0.81) in Dukes’ B and C patients. A significant
progression free survival (PFS) (8 m vs. 8.9 m). proportion recurs with asymptomatic liver
In combination with oxaliplatin/5FU, it increased metastasis and with active surveillance up to
RR (36% vs. 46%) but did not improve PFS. The 20% patients are candidates for curative surgery.
most common toxicities of cetuximab are skin Hence following radical treatment for Dukes’ B
reaction and diarrhoea. or C cancer, patients fit for further active treat-
The K-ras gene is emerging as both a predic- ment are followed up as follows:
tive and prognostic factor for response to tar-
geted anti-EGFR antibodies. Improved response • 6-monthly CEA for 2 years followed by yearly
rate and RR and PFS are associated with wild until 5 years.
type K-ras. Mutated K-ras appears to be associ- • CT liver (and pelvis in rectal cancer) at 12 and
ated with lack of response and worse survival 18–24 months.
compared to wild type K-ras, with some evidence • CT if rising CEA with PET if CT fails to
that anti-EGFR antibodies are actually detrimen- identify relapse.
tal compared to chemotherapy alone.
Screening
Palliation of liver metastases General population
Percutaneous radio-frequency ablation (RFA) A meta-analysis has shown that screening by
can be useful to palliate inoperable or recurrent faecal occult blood (FOB) reduces the risk of
liver metastasis in selected patients (achieves a death from CRC by 16% overall, and by 23%
median survival of up to 36 months). However, (RR 0.77) in those who are actually screened.
lesions >4–5 cm, superficial lesions and those The NHS Bowel Cancer Screening Programme
close to vessels are not effectively treated by RFA. (BCSP) for people aged 60–69 offers 2-yearly
FOB tests and colonoscopy to those with abnor-
mal results. Those >70 can request screening.
Follow-up
Eighty percent of recurrences from CRC occur High-risk populations (p. 50)
within the first 2 years and 90% within 3 years.
5- and 7-year survivals equate to cure for colon Prognosis and outcome
and rectal cancer respectively with <5% relaps- Stage is the most important prognostic factor.
ing after this. Aims of follow-up following radical Stage-wise 5-year survival rates are >90% for
169
II SITE-SPECIFIC CANCER MANAGEMENT
stage I (Dukes’ A), 70–85% for stage II (Dukes’ Investigations and staging
B) without risk factors, 40–60% for stage II with Initial assessment includes digital rectal examina-
risk factors and stage III (Dukes’ C) and 5% for tion, examination under anaesthesia (EUA) and
stage IV disease. Median survival of inoperable biopsy. Transanal US is useful in assessing depth
metastatic disease is 6 months with best support- of invasion and perirectal nodes. CT or MRI is
ive care alone, 18–20 months with chemotherapy useful to assess pelvic nodal metastases. Up to
and over 2 years with targeted agents. 50% of palpable or radiologically suspicious
inguinal nodes are not involved and hence a fine
Anal cancer needle aspiration (FNA) of clinically suspicious
Squamous cell cancers (SCC) of the anal canal nodes, with excision biopsy if FNA is non-diag-
are uncommon. The peak incidence occurs in the nostic, is recommended. Distant metastasis is
seventh decade. There is a slight predominance seen in <5% at diagnosis and staging includes CT
of females of 2–3 : 1 for cancers of the anal chest and abdomen. Apart from routine blood
canal. Risk factors include genital infection with tests, patients with risk factors should be tested
human papillomavirus (HPV, most frequently for HIV.
type 16), chronic immunosuppression in trans- TNM staging is given in Table 11.3.
plant and HIV positive patients, and smoking
Management
(2–5 fold increase).
The treatment options for localized tumours are:
Pathogenesis and pathology • Local excision (selected stage I patients)
The anal canal is 3–4 cm long and extends to the • Concurrent chemoradiotherapy (CRT) (stage
palpable upper border of the anal sphincter. Peri- I–III)
anal cancers by definition occur within 5 cm of • Radical surgery (T3 or 4 causing sphincter
the anal verge with no extension into the canal. destruction and salvage after CRT)
Anal cancer may arise in anal intraepithelial
neoplasia (AIN), which can progress from low to
high grade and is found in areas adjacent to
squamous cell carcinoma. Table 11.3: Staging for anal cancer
Ninety percent of anal cancers are squamous Stage group TNM stage % patients
cell, histological subtypes which include large cell
I T1 (tumour ≤2 cm) N0 12
keratinizing, basaloid, and transitional (large cell
non-keratinizing). Collectively they are referred II T2 (tumour 2–5 cm) N0 55
T3 (tumour >5 cm) N0
to as cloacogenic or epidermoid cancers. All have
a similar natural history, response to treatment, IIIA T1–3 N1 (perirectal 30
nodes)
and prognosis.
T4 (tumour invading
Approximately 5–10% tumours are adenocar- adjacent organ) N0
cinoma of the anal glands and are managed as
IIIB T4N1
low rectal cancers. Other histologic types are Any T N2 (unilateral
melanoma, basal cell and small cell carcinoma. internal iliac or
inguinal nodes)
Presentation Any T N3 (perirectal
Anal carcinoma presents with discomfort, itch and inguinal or
ing and bleeding in 50%. Symptoms are often bilateral internal
iliac +/− inguinal
dismissed as haemorrhoids. Any patient over the
nodes)
age of 60 should be examined to exclude carci-
noma as should those with a mass, enlarged IV M1 (extra pelvic <5
metastases)
inguinal nodes, pain or persistent symptoms.
Faecal incontinence and vaginal fistula are late NB Invasion of rectum, subcutaneous tissue, skin or sphincter
symptoms of locally advanced disease. Distant muscles is not T4
metastases are rare at presentation.
170
CANCERS OF THE GASTROINTESTINAL SYSTEM 11
Treatment options for patients with stage IV alone in terms of reduction in locoregional
disease are palliative chemotherapy, palliative failure, improvement in colostomy free and
radiotherapy and active symptom control. disease specific survival. Acute toxicities are
increased by concurrent treatment, without
Local excision
significant increase in late morbidity.
Local excision with curative intent is an option
for selected patients with stage I disease not Radiation dose and technique
invading the sphincter muscles. The 5-year The dose of radiation concurrent with che
overall survival with local excision alone is motherapy required to treat anal cancer is
approximately 70%. However CRT gives better debatable.
chance of overall disease control and should There is evidence to suggest doses as low as
therefore be considered for all patients who are 30 Gy in combination with chemotherapy are
fit for this approach, even if local excision has sufficient for microscopic disease and 45 Gy
been performed as first line treatment. adequate to treat non bulky gross disease. This
underlies current UK practice. Since the median
Chemoradiation (CRT) potential doubling time of anal cancer is 4 days
There are no RCTs of surgery versus CRT; but (similar to that of cervix cancer), local control
CRT offers sphincter preservation for the major- rate is likely to be decreased by prolonging
ity of patients. Chemoradiation using concurrent overall treatment time.
mitomycin C (MMC) and 5 FU is the standard The failure rate in untreated clinically normal
of care for all stages of locoregional disease (Box inguinal nodes is up to 25%. This has led to the
11.12). The UKCCCR ACT I and an EORTC practice of elective nodal irradiation which reduces
study confirmed the benefit of CRT over RT the risk to <5% without significant morbidity.
The role of cisplatin
Box 11.12: CRT for anal cancer Cisplatin in combination with 5 FU is under
Radiotherapy
investigation as neoadjuvant, concurrent and
Phase I adjuvant treatment in anal cancer. The current
Patient prone with anal marker (canal tumours) and UK–ACT II is comparing two CRT schedules
palpable disease wired (margin tumours and (standard MMC/5FU versus cisplatin/5FU) and
inguinal nodes)
adjuvant chemotherapy with two cycles of
Ant and post fields
cisplatin/5FU versus no further treatment.
Borders
Superior = 2 cm above bottom of SI joints HIV positive patients
Inferior = 3 cm below anal margin or most inferior Prior to the advent of highly active antiretroviral
extent of disease
therapy (HAART), tolerance of CRT in patients
Lateral = lateral to femoral heads
with HIV and CD4 counts <200 at doses of RT
Dose 30.6 Gy in 17 fractions of 1.8 Gy
>30 Gy was poor, with excessive toxicities. The
Phase II majority of patients can now be treated with
Field borders are a 3 cm margin on GTV in all planes.
standard doses (p. 297).
N0 canal tumours = 3 field (posterior, right and left
lateral)
Radical surgery
N0 margin tumours = direct field
N+ tumours = anterior and posterior fields Patients with confirmed local relapse following
Dose 19.8 Gy in 11 fractions of 1.8 Gy CRT who are fit to consider salvage surgery
should be restaged with CT chest and abdomen
Chemotherapy
Week 1 mitomycin C 12 mg/m2 d1 and 5FU 1000 mg/ to exclude metastatic disease as well as MRI
m2/d d1–4 pelvis to stage regional disease. Approximately
Week 5 5FU 1000 mg/m2/d d1–4 50% can be cured by a salvage surgical resec-
tion, but local recurrence rates approach 50%
RT fields and doses are as per ACT II protocol. Chemotherapy
schedule is as per ACT I study even in patients undergoing complete resections.
Inguinal node dissection similarly is reserved for
171
II SITE-SPECIFIC CANCER MANAGEMENT
172
Cancers of the
genitourinary system
M Beresford
12
Cancer of the kidney • Acquired cystic kidney disease – in dialysis
patients.
Introduction • Genetic factors – Von Hippel–Lindau disease
is a familial disorder characterized by the
Renal cell carcinoma (RCC) accounts for 2–3% development of multiple tumours including
of all cancers, with 6600 new diagnoses and bilateral renal cell carcinomas, retinal/central
3600 deaths per year in the UK. There has been nervous system haemangioblastomas and
an increasing incidence over the past 10–15 phaeochromocytomas. Hereditary papillary
years, at least in part due to more incidental renal cell carcinoma (HPRC) is a condition in
tumours being found with the increased use of which patients are at risk of developing mul-
cross-sectional imaging. Peak age at presentation tiple, bilateral type 1 papillary renal carcino-
is between 65–75 years, with very few cases in mas and involves mutation in the c-met
the under 40 age-group. The male to female ratio oncogene.
is 3 : 2.
Pathology
Aetiology In adults, 85% of malignant lesions arising from
Risk factors for renal carcinoma are as follows: the kidney are renal cell carcinoma, with most
of the remainder being metastases from other
• Smoking – smokers are more likely to develop
primary tumours. Other rarer cancers include
RCC than non-smokers and the effect seems
clear cell sarcoma, lymphoma, oncocytoma and
to be dose-dependent, with heavy smokers
adult form of Wilms’ tumour.
having a relative risk of 2.0.
• Occupational exposure – risk ratios of 1.3 to • Renal cell carcinomas (RCCs) – most RCCs
1.6 have been observed in individuals exposed are well-differentiated and usually arise within
to petroleum, hydrocarbon, steel, asbestos, the upper pole. The majority are of clear cell
cadmium or dry cleaning products. origin although they often contain several dif-
• Analgesic nephropathy – there is an increased ferent cell types, including granular cells and
risk of RCC in patients who develop nephro spindle cells (the poor prognosis sarcomatoid
pathy associated with the use of phenacetin- variant).
containing analgesics. • Transitional cell carcinomas – are relatively
• Obesity – obesity increases the risk of devel- uncommon, accounting for 5% of malignan-
oping RCC, with a linear relationship between cies in the kidney, and are sometimes a result
body weight and risk (odds ratios of up to of analgesic nephropathy. They arise in the
4.8). Excessive consumption of red meat and renal pelvis and often affect multiple sites of
dairy products might increase risk, but there the urothelial mucosa with 50% having a
appears to be no relationship with alcohol or history of bladder tumours. Transitional cell
caffeine intake. carcinomas of the renal pelvis are usually
Presentation
Figure 12.1
Renal tumours often remain asymptomatic and
CT scan showing an irregular mass lesion arising from
unnoticed until large. The classical triad (also the left kidney.
known as ‘too late’ triad) of presenting symp-
toms (loin pain, haematuria and flank mass)
occurs in less than 10% of patients. Up to 20%
Staging
of patients have a history of fever at presentation,
and a third report weight loss and cachexia. 30% Stage (Table 12.1) determines the prognosis and
have metastatic disease at presentation and a treatment of renal tumours.
further 25% have locally advanced disease.
Management
Typical sites of metastases include lungs (75%),
lymph nodes/soft tissue (36%), bone (20%) and Surgery of the primary tumour
liver (18%). Patients may also demonstrate a Curative
number of paraneoplastic manifestations such as The definitive treatment for renal cancer is surgi-
polycythaemia, hypercalcaemia etc. cal resection. Radical nephrectomy is performed
by an anterior approach and en bloc removal of
Investigations and staging the kidney, adrenal and perirenal fat. The adrenal
Initial investigations include urinalysis for gland is rarely involved in T1 or T2 tumours and
protein, blood and cytology. Blood tests should can be left behind. If tumour invades the renal
include full blood count, urea and electrolytes, vein, this can be ligated distal to the tumour
liver function, clotting and calcium levels. thrombus and even partial vena caval resection
Imaging may be undertaken if necessary.
Regional lymphadenectomy is sometimes per-
CT scan: CT scan appearance (Figure 12.1) is formed during the radical procedure, but its role
important in determining the appropriate surgi- in prolonging survival is debatable.
cal procedure – laparoscopic, partial or radical Recent improvements in surgical techniques
nephrectomy. Size is important in diagnosis – have enabled laparoscopic partial or total
benign cortical adenomas can look like malignan- nephrectomies as a less invasive technique for
cies but tend to be less than 3 cm in diameter. removing kidneys with small volume tumours.
Other investigations: for small tumours, a Nephron-sparing surgery should be considered
chest X-ray is sufficient for staging if the CT scan particularly for patients with poor renal reserve
does not include the chest. Ultrasound and colour or non-functioning contralateral kidneys.
flow Doppler are useful in assessing renal vein
and inferior vena caval invasion. A bone scan Palliative
should be considered if there are symptoms or a Nephrectomy should also be considered as a
raised alkaline phosphatase. palliative procedure in patients with metastatic
If there are concerns about renal reserve disease. Although very rarely (<1%) this may
from imaging or renal function blood tests, result in regression of metastases (‘abscopal
then more formal assessment of renal function is effect’), it is not the main reason for considering
174 appropriate. surgery. It is aimed mainly at reducing symptoms
CANCERS OF THE GENITOURINARY SYSTEM 12
such as pain, haematuria and hypercalcaemia. The optimal regimen of interferon is not yet
Removal of the tumour bulk may also improve known. Typical dose schedules are of 9–12 mega-
the efficacy of any subsequent systemic treatment units subcutaneously three times per week, often
– trials have shown that patients with metastatic starting at a lower dose to assess tolerability.
disease who are treated with interferon have Treatment is discontinued at disease progression
improved survival if they undergo palliative or after 6–9 months, whichever comes first.
nephrectomy prior to the immunotherapy. Blood counts and liver function should be moni-
tored during treatment.
Adjuvant therapy
Interferon has significant side effects (Box
Adjuvant therapy with interferon-α following 12.1) that impact on quality of life. These effects
nephrectomy has been investigated and does are dose dependent, but the majority tend to stop
not appear to confer any survival advantage. quickly on discontinuation of treatment. Common
More complicated and toxic regimes using com side effects include fatigue, flu-like symptoms,
binations of interferon, interleukin-2 and 5- diarrhoea, nausea and vomiting, anorexia, bone
fluorouracil (known as the Atzpodien regimen) marrow suppression and rash at injection site.
have not proved any better and in fact may be
detrimental. Interleukin 2
Interleukin works by stimulating cytotoxic
Metastatic disease
T-cells. It can be given subcutaneously, by high-
Interferon-alpha dose bolus intravenous injection or by continu-
Interferon alpha is a cytokine with anticancer and ous infusion (all give similar results). It has been
antiviral activity. Response rates in metastatic used instead of or in combination with inter-
RCC are reported as 10–15% with a 2% com- feron, and seems to result in improved response
plete response rate (although there are also occa- rates (of 15–20%), but with no improvement in
sional reports of spontaneous remissions off overall survival and significantly worse toxicity
treatment) and a stable disease rate of approxi- than interferon.
mately 25%. A Cochrane review showed a
3-month survival benefit with interferon com- Medroxyprogesterone-acetate
pared with no interferon. Interferon may be better The response rate is only 5–7%. Trials compar-
in ‘good’ prognosis metastatic disease, based on ing progestins and interferon therapy show a
the number of disease sites, performance status survival benefit of approximately 2.5 months in
and diagnosis to metastases interval. favour of the interferon. 175
II SITE-SPECIFIC CANCER MANAGEMENT
Cancers of the urinary bladder carcinoma with tumours occurring in the ureters
and renal pelvis as well as the urinary bladder.
and renal pelvis Carcinoma-in-situ manifests as severely dys-
plastic cells through all layers of the epithelium.
Introduction It carries a poor prognosis and often has an inva-
Bladder cancer accounts for 4% of all malignan- sive component. At least 30% of grade III carci-
cies, with over 10,000 new diagnoses and almost noma-in-situ will become invasive.
5000 deaths each year in the UK. Worldwide it
is the 9th most common cancer, but is more Presentation
common in North America and Western Europe, The most common presentation is with painless
where it is the 4th most common cancer in males haematuria. A patient presenting with macro-
and 8th most common in females. The world- scopic haematuria has a 25% chance of having
wide male to female ratio is 10 : 3. Bladder cancer a bladder tumour (5% if microscopic haematu-
is rare in the under 50 age group, and most ria). Other symptoms include frequency, urgency,
common in over 70-year-olds. dysuria and loin pain due to ureteric obstruction.
Occasionally patients present with symptoms
Aetiology caused by metastases such as bone pain, fractures
Risk factors for bladder tumours include the or shortness of breath.
following: Lymph node involvement is strongly related to
• Smoking (2–5 fold increased risk). tumour grade. Less than 10% of grade 1 tumours
• Infections – especially schistosomiasis have nodal involvement, compared with 80% of
which increases the risk of squamous cell grade 3. Metastatic spread is predominantly to
carcinoma. the lungs and bones.
• Occupational/chemical (aniline dyes, aro-
Investigations and staging
matic amines, printing industry, rubber
industry, firelighter manufacturers, tar manu- Investigations
facturing, sewage workers, pest control, Initial investigations include urine cytology,
benzidine, cyclophosphamide, 2-naphthyline, intravenous urogram (IVU) and cystoscopy. Cys-
xenylamine, phenacetin). toscopy should include bimanual examination,
• Bladder calculi (chronic cystitis). complete inspection of the urethra and bladder,
• Chronic indwelling catheters. biopsies of all suspicious areas, transurethral
• Previous radiotherapy (risk of sarcoma). resection of visible tumour and a detailed diagram
of the findings to assist in comparison at follow-
Pathology up procedures.
In Western countries, over 90% of bladder Once a bladder tumour is confirmed further
tumours are predominantly transitional cell car- investigations should be considered, including
cinoma (TCC), often with elements of squamous, blood tests (renal function, full blood count,
adenocarcinoma or sarcomatous differentiation. liver function and calcium level), chest X-ray,
80% are papillary and 20% flat in appearance histology review and for muscle-invasive tumours,
and the most likely areas affected are the poste- CT or MRI scan of the abdomen and pelvis
rior and lateral bladder walls. Flat tumours tend (Figure 12.2). MRI is better for showing early
to be of higher grade and more aggressive. Less invasion of adjacent organs and extravesical
common histological types are squamous cell spread. A bone scan is only necessary if indicated
carcinoma, adenocarcinoma and very rarely by symptoms or a raised alkaline phosphatase.
small cell carcinoma. Chest X-ray or CT chest is useful to rule out
Tumours often present initially as low grade metastatic disease (Figure 12.3).
and superficial, but tend to be multiple and recur-
rent. With each recurrence there is a tendency to Staging
progression in cytologic atypia. The whole of the The staging of bladder tumours is shown in
uroepithelial tract is at risk of transitional cell Table 12.2.
177
II SITE-SPECIFIC CANCER MANAGEMENT
A B
Figure 12.2
CT scan of bladder tumour: tumour shown as filling defect in the right side of base of bladder (short arrow) with
contrast filled ureter (long arrow) entering the bladder through the tumour (A). Scan shows an irregular mass arising
from base of the bladder (B).
Tis Ta T1 T2 T3 T4
Transurethral
resection
If no response,
consider cystectomy
Figure 12.4
Management of transitional all bladder carcinoma by T stage.
mitomycin-C is being investigated as a less toxic published data as to the benefits of this approach.
regime to combine with radiotherapy. The current evidence for adjuvant chemo
therapy is stronger for node positive disease than
Management of carcinomas of for locally advanced but node negative disease.
the renal pelvis and ureter Metastatic disease is treated with similar plat-
The staging of tumours of the renal pelvis inum-based chemotherapy regimes to those used
and ureter is given in Table 12.3. Renal pelvic in metastatic bladder cancer.
and upper urothelial transitional carcinomas are
treated by radical resection of the kidney and Management of small cell carcinoma of
ureter. Nephron-sparing procedures can be the bladder
undertaken in small, localized tumours if there Small cell bladder cancer accounts for less than
are concerns about function of the remaining 1% of bladder cancers and commonly presents
kidney. Adjuvant radiotherapy confers no sur- at an advanced stage. In localized disease patients
vival advantage following complete resection, should be treated with neoadjuvant platinum-
but may be considered in patients with positive based chemotherapy prior to surgery or radio-
margins or residual local disease. Doses are therapy. Responses to chemotherapy are seen in
limited by surrounding structures, but typically metastatic disease, but median survival is less
45–50.4 Gy in 25–28 fractions can be delivered. than 12 months. Most published data relates to
There is some evidence to suggest that the addi- the use of neuroendocrine regimes such as carbo-
tion of concurrent cisplatin chemotherapy to platin/cisplatin with etoposide.
radiotherapy improves overall and disease-free
survival in T3/4 and/or node-positive disease Survival
following surgical resection. The majority of 5-year survival rates for bladder cancer are as
relapses are distant, so systemic adjuvant therapy follows:
using platinum-containing chemotherapy regimes
Stage 5-year survival
should be considered, although there is little
Ta, T1, CIS 70–95%
T2 50–80%
T3a 40–70%
T3b 20–50%
T4 10%
Table 12.3: Staging of tumours of the renal
pelvis and ureter
Stage Survival with metastatic disease is poor despite
I T1 N0 M0 Tumour invades subepithelial reasonable response rates to chemotherapy, with
connective tissue <10% 2-year survival.
II T2 N0 M0 Tumour invades muscularis
III T3 N0 M0 Tumour invades peripelvic (or Cancer of the prostate
periureteric) fat or renal
parenchyma Introduction
IV T4 T4 Invades adjacent organs or Prostate cancer accounts for 12% of all cancers
perinephric fat
(23% of cancer in men), with 32,000 new diag-
or N1–3 N1 Metastases in single node noses and 10,000 deaths each year in the UK.
≤2 cm There has been a large increase in incidence over
N2 Metastases in single node
>2 cm but ≤5 cm or multiple
the past 10–15 years due to increased detection
nodes through PSA screening and surgery for benign
N3 Metastases in single node prostatic disease. The majority of cases occur in
>5 cm the over 70 age-group and the disease is rare in
or M1 M1 Distant mets the under 50s. As yet there is no evidence that
PSA screening reduces mortality rates from pros-
tate cancer.
182
CANCERS OF THE GENITOURINARY SYSTEM 12
>10–15 ng/ml). If there is doubt over lymph node time, expressed in months or years, and the
involvement, a laparoscopic retroperitoneal PSA velocity, expressed as ng/ml/year (this is
lymph node biopsy might be considered prior to commonly utilized in patients on active sur-
proposed radical treatment. veillance – see below). To get an accurate
Table 12.4 shows the staging of prostate indication of PSA velocity, at least three meas-
cancer. urements should be taken over a period of 18
As well as the absolute PSA level, various months.
other PSA parameters have been developed in an Note that very poorly differentiated cancers
attempt to improve the predictive value of the may not secrete PSA and are therefore more dif-
test for diagnosis and monitoring: ficult to diagnose, predict and monitor.
• PSA density (PSA/volume of gland) allows for
higher PSA levels in older men with large,
Management of localized disease
hypertrophied glands. A value of 0.1 ng/ml/cc The treatment options for localized disease are:
is considered normal (e.g. a PSA of 5 ng/ml • Active surveillance (for patients suitable for
with a 50 cc prostate volume). radical treatment).
• Percent-free PSA (fPSA) is the ratio of how • Watchful waiting (for patients not suitable for
much PSA circulates free compared with the radical treatment).
total PSA level, including that attached to • Radical treatment with surgery or radiother-
blood proteins. The percentage of free PSA is apy or both.
lower in men who have prostate cancer than
in men who do not and may be useful in Active surveillance and watchful waiting
determining which patients with intermediate Many patients will have slow-growing disease
PSA levels should have a prostate biopsy for which may have little or no impact on their life-
diagnosis. There is some controversy over expectancy, and they might therefore be spared
where the cut-off should lie, but fPSA levels the toxicities and inconvenience of radical treat-
of <10% are very suspicious. ment. Active surveillance implies close monitor-
• PSA kinetics give an indication of the rate of ing with early curative treatment offered to
184 tumour growth, and include the PSA doubling patients who show signs of progression. There
CANCERS OF THE GENITOURINARY SYSTEM 12
are no absolute criteria for considering active Box 12.5: Radiotherapy for prostate
surveillance and the decision should be made (see Figure 12.5)
with the patient, but typical parameters would Localization
be T1–T2b disease, Gleason grade ≤7, PSA ≤15 CT planning with empty rectum. Target volume
(with favourable kinetics). A surveillance pro- defined on planning CT or co-registered CT/MRI
gramme might consist of 3-monthly visits for the Target volume
first 2 years, followed by 6-monthly visits there- Primary radical treatment
after, with a DRE and PSA checked at each visit. CTV = Whole prostate + tumour extension
Repeat transrectal biopsies should be performed Whole seminal vesicles included if risk of involvement
≥15%
at 18 months. Criteria for consideration of
Consider treating pelvic nodes if risk of nodal
radical treatment would be PSA progression involvement ≥15%
(doubling time <2 years), clinical progression or Phase I PTV = CTV + 1 cm margin
upgrading of the Gleason score on repeat biopsy. Phase II PTV = CTV + 0.5 cm margin
‘Watchful waiting’ is appropriate where Salvage radiotherapy
patients deemed unsuitable for radical treatment Prostate bed, any surgical clips and residual seminal
are treated with palliative endocrine therapy vesicle with 1 cm margin
when there is symptomatic progression. Dose
Prostate and seminal vesicle
Radical treatment
Phase I – 56 Gy in 28 fractions
Radical treatment options for prostate cancer Phase II – 18 Gy in 9 fractions
include radical prostatectomy, external beam Whole pelvis and prostate
radiotherapy and brachytherapy (low-dose rate Phase I (whole pelvis with prostate) – 50 Gy in 25
or high-dose rate). Each treatment has its own fractions
characteristics and may be suitable for certain Phase II (prostate alone) – 18–24 Gy in 9–12
types of patients, but reported success rates are fractions
Salvage radiotherapy
similar if patients are chosen appropriately.
66–70 Gy in 33–35 fractions
Radical prostatectomy Tolerance
Perineal, retropubic or laparoscopic approaches • Rectum: 55.5 Gy to no more than 50% of the
can be considered. There is a 5–15% risk of rectum, 70 Gy to no more than 25% and 74 Gy
to no more than 3%.
urinary dysfunction after surgery. Nerve-sparing
• Bladder: Less than 50% of the bladder should
techniques have improved morbidity, with ap- receive 67 Gy.
proximately 50% impotence rates.
External beam radiotherapy
Conformal CT planning is well established in (1.2–1.5 Gy), which implies that hypofraction-
prostate radiotherapy (Box 12.5 and Figure ated courses of radiotherapy with a high dose per
12.5). The Medical Research Council RT01 fraction might result in improved cancer control
study randomized between 64 Gy and 74 Gy and for a similar level of side effects. For this reason
found a hazard ratio for biochemical progression doses such as 57–60 Gy in 19–20 fractions over
free survival of 0.67 (CI 0.53–0.85; p = 0.0007) 4 weeks are being investigated. These shortened
in favour of the escalated group. However this schedules have the additional advantage of
was achieved at the expense of increased late sparing resources and being more convenient for
bowel and bladder toxicity. It is not yet known patients.
whether there will be any improvements in Though the survival benefit of whole pelvic
overall survival. radiotherapy in high risk of pelvic lymph node
Newer techniques such as intensity modulated involvement (≥15% on Roach criteria) remains
radiotherapy (IMRT) might enable even further to be demonstrated, it is commonly practised.
increases in the dose administered or further Acute side effects of radiotherapy include
sparing of normal tissues. dysuria, frequency, diarrhoea, lethargy and ery-
Radiobiological studies have suggested a sur- thema. Late effects include proctitis (diarrhoea,
prisingly low alpha-beta ratio for prostate cancer rectal bleeding, tenesmus: 30% mild, 5% severe), 185
II SITE-SPECIFIC CANCER MANAGEMENT
Figure 12.5
Target volumes for radical prostate
radiotherapy. Orange line phase I
PTV including prostate and seminal
vesicle with a 10 mm margin and
purple line phase II PTV including
only prostate with a 5 mm margin.
impotence (30–40%) and urinary incontinence following a shortened course of external beam
(1–5%). radiotherapy but can also be used as mono-
therapy. Treatment is delivered through catheters
Brachytherapy implanted with ultrasound guidance into the
Low-dose rate (LDR) brachytherapy prostate under general anaesthetic. HDR boost
Permanent radioactive seeds are implanted patients are typically treated in two or three frac-
directly into the prostate via transperineal needles tions, 12 hours apart, necessitating overnight
that are inserted with ultrasound guidance under stay with the catheters and template in-situ. Box
general or spinal anaesthetic. Approximately 12.6 shows some of the HDR boost schedules
50–100 Iodine-125 or Palladium-103 seeds are currently in use.
implanted to achieve a prescribed dose of 145 Gy.
Patients eligible for this treatment are those with Role of hormones in curative treatment
a prostate volume of <50 cc, Gleason ≤6, PSA Patients undergoing radical radiotherapy are
≤15, T2 or less disease, those haven’t had a previ- commonly treated with 3 months of neoadjuvant
ous TURP and not at high risk of extracapsular luteinizing hormone releasing hormone (LHRH)
extension or lymph node involvement. Patients analogues. This approach may enable a reduc-
should have a transrectal ultrasound volume tion in the volume of tissue irradiated due to
study to assess suitability for brachytherapy. shrinkage of the prostate gland. An EORTC
Although there is good evidence to confirm the study compared radiotherapy alone versus radio-
efficacy of brachytherapy in terms of PSA control therapy with immediate androgen suppression
and biopsy findings, there is as yet no long-term started on the first day of radiotherapy and con-
survival data. The procedure has not been directly tinued for 3 years. 5-year clinical disease free
compared with external beam radiotherapy survival was 40% versus 74% and overall sur-
or radical surgery in randomized studies, but vival was 62% versus 78% in favour of the adju-
comparative and cohort studies show similar vant endocrine group.
5-year biochemical recurrence-free and overall Prolonged adjuvant treatment for 2–3 years
survival. should be offered to all patients with high risk
disease (Gleason 8–10, clinical T3/4 tumours or
High-dose rate (HDR) brachytherapy lymph node risk >30%). The role in low and
HDR brachytherapy is suitable for intermediate- intermediate risk patients is being assessed in
high risk patients. It is typically given as a boost randomized studies, but these patients are cur-
186
CANCERS OF THE GENITOURINARY SYSTEM 12
Box 12.6: High-dose rate brachytherapy Box 12.7: Side effects of LHRH analogues
boost schedules
Hot flushes
External beam dose HDR brachytherapy boost dose Weakness/loss of muscle bulk
and fractionation and fractionation
Weight gain
35.7 Gy in 13 17 Gy in 2
Fatigue
46 Gy in 23 16.5 Gy in 3
46 Gy in 23 17 Gy in 2 Osteoporosis/fracture risk
50 Gy in 25 18 Gy in 2 Loss of libido and erectile function
45 Gy in 23 16.5 Gy in 3
Mood changes
46 Gy in 23 23 Gy in 2
Poor concentration/memory
rently treated with 3–6 months of LHRH ana- Metastatic disease, elderly and those with
logues before and during radiotherapy. significant comorbidities
It should be noted that LHRH analogues Metastatic disease (Figure 12.6) is generally
are not without side effects (see Box 12.7) and treated with endocrine therapy, usually LHRH
can add considerably to the morbidity of patients analogues in the first instance, with the addition
undergoing radiotherapy. of an anti-androgen to give maximal androgen
There is no established role for neoadjuvant blockade on biochemical or clinical progression.
or adjuvant hormonal manipulation in patients The typical duration of response to first line endo-
undergoing radical prostatectomy. crine therapy with LHRH analogues is 18–24
Management of locally advanced and months. There is increasing interest in intermit-
tent LHRH analogue therapy as a way of pro-
metastatic disease
longing the useful lifespan of the drug and of
Locally advanced disease allowing patients to have periods of time off
For patients with locally advanced disease radical treatment and therefore avoid some of the side
radiotherapy or surgery may add little or no effects. Typical schedules are to treat until PSA
additional benefit over hormone therapy alone, falls below 4 ng/ml (or <80% of initial value) and
although there is emerging evidence that radio- restart when PSA rises above 10 ng/ml. Studies
therapy does improve biochemical control in have shown no difference in survival when com-
patients with PSA levels up to 70 ng/ml. The pared with continuous therapy, but significant
mainstay of treatment is with LHRH analogues improvements in quality of life, with patients
or anti-androgens. Prolonged treatment with spending a median of 1 year off treatment.
LHRH analogues results in significant toxicity Third line hormonal therapy with oestrogens
due to reduction of testosterone levels (see Box can be considered but response duration tends to
12.7). Androgen receptor inhibitors, such as be short unless response to first and second line
bicalutamide, or 5-alpha reductase inhibitors treatment has been particularly good.
effectively reduce the delivery of testosterone to Elderly patients and those with significant co-
the prostate without reducing serum testosterone morbidities can be treated symptomatically with
levels and therefore tend to have a better side or without hormones.
effect profile, although they can cause significant
gynaecomastia. Prophylactic radiotherapy to the Management of relapse
breast buds or prophylactic tamoxifen 10–20 mg/ after radical treatment
day can reduce and sometimes prevent the painful Some authorities advocate postoperative radio-
gynaecomastia. Typical radiotherapy doses are therapy to the prostatic bed in cases with a posi-
8–10 Gy in a single fraction or 15 Gy in three tive margin at radical prostatectomy, whilst
fractions given on alternate days, using electron others would wait and offer radiotherapy if the
radiotherapy delivered to an 8–10 cm circle PSA fails to completely suppress or subsequently
around each nipple. rises. In these situations, salvage radiotherapy is
187
II SITE-SPECIFIC CANCER MANAGEMENT
Figure 12.6
Bone scan in metastatic prostate
cancer. Note the absence of
uptake in the kidneys and bladder
(arrows) suggesting that uptake
is preferentially by the extensive
metastases. This picture is
called a ‘superscan’ which is a
contraindication for radioisotope
treatment (risk of lethal bone
marrow suppression)
most effective if given before the PSA reaches a weeks) when compared with the previous stand-
value of 2 ng/ml (Box 12.5). ard regimen of mitoxantrone and prednisolone.
Salvage surgery for radiotherapy failures is Doses of 75 mg/m2 are administered on a 3-weekly
rarely undertaken due to the complicated nature basis for up to 10 cycles. Although low-grade
of the procedure and questionable results. Newer neutropenia is fairly common, the rates of febrile
invasive techniques such as cryotherapy and high neutropenic sepsis are reassuringly low (<3%)
frequency ultrasound (HiFU) have shown some without colony-stimulating factor support.
success in terms of PSA control after radiother- Chemotherapy agents are now being investi-
apy failures (and perhaps as primary treatments gated in earlier stages of prostate cancer, both in
in place of radiotherapy), but long-term out- metastatic disease prior to the development of
comes are awaited. CRPC or as an adjuvant to radical prostatectomy
for localized disease. There is also interest in
Role of chemotherapy second-line chemotherapy after failure of taxanes;
in prostate cancer there is some evidence for PSA response with
Until recently, chemotherapy has been reserved satraplatin, an orally active platinum-based drug.
for advanced metastatic prostate that has become Cabazitaxel, a new taxane, has demonstrated a
refractory to endocrine treatment (known as 2.5 month survival benefit in the second-line
hormone refractory prostate cancer, HRPC, or setting.
more accurately as castration resistant prostate
cancer, CRPC). Docetaxel is now well established Palliative treatment
in this setting, with studies showing quality of life • Steroids: Low-dose steroids (0.5–2 mg of
and overall survival benefits (in the order of 10–12 dexamethasone daily) are effective in some
188
CANCERS OF THE GENITOURINARY SYSTEM 12
patients in terms of both quality of life men, with around half in men under 35 years of
improvements and PSA control. age and 90% in men under 55 years.
• Bisphosphonates: Zoledronic acid adminis-
tered intravenously once a month has been Aetiology
shown to reduce the incidence of skeletal Apart from age and race, other risk factors
events, including fractures or the need for include a previous history of testicular cancer or
radiotherapy, in men with bone metastases. It carcinoma-in-situ. Cryptorchidism (undescended
is effective at controlling refractory metastatic testes at birth) increases the risk by 2–4 fold.
bone pain. There are some reports of increased incidence in
• Radiotherapy: External beam radiotherapy is men with subfertility and also suggested links
very effective in controlling metastatic bone with high dietary dairy product intake and a
pain and symptoms of primary tumour inva- sedentary lifestyle.
sion such as haematuria and pain. Prolonged
fractionation regimes have not been shown to Pathology
be more beneficial than a single fraction of The majority of testicular malignancies are germ
8 Gy. Radiotherapy is also commonly used in cell tumours (>90%), either seminomas or ter-
cases of spinal cord or nerve root compression atomas. Approximately 20% are of mixed cell
from vertebral metastases (p. 328). type and should be classified as non-seminoma-
• Radioisotopes: Strontium-89 and Samar- tous germ cell tumours (NSGCT). Other types
ium-153 are effective at controlling pain in up include sex cord tumours, lymphomas and
to 80% of patients with widespread bone sarcomas.
metastases. Radioisotope treatment is partic- Intratubular germ cell neoplasia has features
ularly useful in patients with diffuse bone similar to carcinoma-in-situ, seen in the contra
pain that cannot easily be targeted with exter- lateral testes in 5% of testicular cancer patients.
nal beam radiotherapy (Figure 12.6). Care There is a risk of progression to malignancy of
must be taken if chemotherapy is to be con- 50% at 5 years. This risk can be prevented with
sidered at a later date because of a suppressive radiotherapy (20 Gy in 10 fractions), which will
effect on the bone marrow. result in infertility, but avoids the need for a
second orchidectomy. It is diagnosed by open or
Prognostic factors and survival needle biopsies and is more likely in young
Survival figures from prostate cancer vary widely, patients with an atrophic contralateral testis.
depending on histology, stage, PSA level and
therapeutic intervention. Patients with localized
Presentation
disease treated with either radiotherapy or radical The majority of testicular tumours (75%) present
surgery have 5-year biochemical control rates of with a painless, firm swelling of the testis. Occa-
75–85% and 10-year overall survival rates of sionally patients present with lower back pain
60–70%. Patients with metastatic disease can due to intra-abdominal or pelvic lymph node
survive for many years, particularly if the tumours spread, or indeed palpable lymphadenopathy,
are hormone-responsive and if the metastatic particularly in the left supraclavicular fossa.
spread is confined to the bones. Other methods of presentation include infertility,
gynaecomastia, secondary hydrocoeles or with
symptoms due to secondary spread to other
Cancer of the testes organs such as lung or liver.
Table 12.6: International Germ Cell Consensus Classification prognostic criteria for non-
seminomatous tumours
Good prognosis Intermediate prognosis Poor prognosis
AFP <1000 ng/ml AFP 1000–10,000 ng/ml AFP >10,000 ng/ml
β-HCG <5000 iu/l HCG 5000–50,000 iu/l HCG >50,000 iu/l
LDH <1.5 times the upper limit of LDH 1.5–10 × ULN LDH >10 × ULN
normal (ULN)
Testis or retroperitoneal primary Testis or retroperitoneal primary Mediastinal primary site
No non-pulmonary visceral metastases No non-pulmonary visceral Non-pulmonary visceral metastases
metastases
190
CANCERS OF THE GENITOURINARY SYSTEM 12
Seminoma
Stage I Stage IIA and IIB Stage IIB Good prognosis Intermediate prognosis
(2–2.5 cm) (2.5–5 cm) IIC/III IIC/III
OR OR
Active treatment–options BEP x 3 BEP x 3 BEP x 3 BEP x 4
Carboplatin 1 course AUC7 or or or
Paraaortic radiotherapy EP x 4 EP x 4 EP x 4
OR
PET PET
(optional
If previous If previous
chemotherapy radiotherapy
Progression
Bold lettering indicates standard treatment Options – salvage chemotherapy, localized RT, local re-irradiation
Figure 12.7
Postoperative management of seminoma.
treatment options are a single dose of carboplatin temic treatment. Standard treatment for stage IA
(AUC7) and para-aortic radiotherapy (20 Gy in and stage IB with 2–2.5 cm node is radiotherapy
10 fractions) (Box 12.9). to para-aortic and ipsilateral iliac nodes (‘dog leg
field’). An alternative option is three cycles of
Stage II–IV disease PEB chemotherapy (3 or 5-day schedule). Treat-
Clinical stage IIA disease should be verified ment option of stage IIB with 2.5–5 cm node is
beyond imaging (e.g. needle biopsy) before sys- three courses of PEB (Box 12.8).
191
II SITE-SPECIFIC CANCER MANAGEMENT
Non-seminoma
Marker+ Marker–
Marker normal: Marker not Marker not normal
Salvage NS-RPLND no residual normal, potentially and irresectable or
therapy tumour resectable residual multiple residual
tumour
Resection Salvage
chemotherapy
Figure 12.9
Postoperative management of non-seminoma.
193
II SITE-SPECIFIC CANCER MANAGEMENT
effusion, although some effusions may be reac- Box 13.1: Risk of malignancy index (RMI)
tive. Haematogenous spread to the liver or lung
Feature RMI score
is unusual (2–3%).
Ultrasound features: 0 = none
multilocular cyst 1 = one abnormality
Presentation solid areas 3 = two or more
Overall 95% of patients diagnosed with ovarian bilateral lesions abnormalities
cancer are symptomatic. The most common ascites
intra-abdominal metastases
symptoms are abdominal distension (61%), Premenopausal 1
abdominal bloating (57%) or pain (36%), indi- Postmenopausal 3
gestion (31%), pelvic pain (26%), constipation CA-125 U/ml
(24%) and urinary incontinence (24%), back
RMI score = ultrasound score × menopausal score × CA-125 level
pain (23%) and dyspareunia (17%). Half of in U/ml.
patients experience constitutional symptoms
(anorexia, weight loss or nausea). The median
duration of symptoms is 3–6 months. CA19-9 (p. 287). The immunohistochemical
Signs profile of an ovarian tumour is typically CK 7
positive and CK 20 negative. A serum CA125:CEA
Physical findings are rare in patients with early ratio >25 is strongly suggestive of an ovarian
disease. Patients with advanced disease may rather than a GI primary.
present with abdominal distension (ascites or
tumour), cachexia, bilateral swollen legs (low Tissue diagnosis
albumin, or large pelvic masses) signs of pleural Patients in whom neo-adjuvant chemotherapy is
effusion which is more often right sided, and indicated should have a tissue diagnosis prior to
pelvic or abdominal mass. surgery, usually by ultrasound or CT guided
biopsy.
Investigations and staging
All patients with suspected ovarian cancer should Preoperative investigations
have estimation of serum CA-125 and ultrasound In addition to imaging and tumour markers,
scan of the abdomen +/− transvaginal ultrasound patients should have full blood count and bio-
scan. The CA-125 is raised in 50% of patients with chemistry for renal and hepatic function. Patients
stage I disease, and in 90% of stage II–IV. Muci- who may require bowel surgery should be
nous tumours often have a normal CA-125. Raised referred to the stoma team for discussion
CA-125 is not specific to ovarian cancer. Young pre-operatively.
patients should also have estimation of germ cell
Staging
tumour markers (AFP, betaHCG, and LDH).
The risk of malignancy index (RMI) scoring The International Federation of Gynaecology
system is used to predict whether a pelvic mass is and Obstetrics (FIGO) staging is shown in Box
malignant (Box 13.1). Women with an RMI of 13.2.
>200 should be referred to a specialist centre for
Management (Figure 13.2)
further management and surgery. An RMI of >200
has a positive predictive value of 87% and a sen- Primary surgery
sitivity of 88% for diagnosing malignant disease. Surgery involves total abdominal hysterectomy
To assess operability patients should have a (TAH), bilateral salpingo-oophorectomy (BSO),
CT of the abdomen and pelvis (Figure 13.1), and infra-colic omentectomy, peritoneal washings,
a CXR. In patients with a pleural effusion, cyto- biopsy of any lesions or adhesions, blind perito-
logical diagnosis is required to determine if the neal biopsies of the bladder, cul de sac, paracolic
effusion is malignant. gutter, hemidiaphragm and pelvic sidewall and
Patients with ascites without a mass on CT, pelvic and para-aortic node sampling. Surgical
should have cytological and immunohistochemi- staging is important to guide treatment with
cal analysis, in addition to CA-125, CEA and chemotherapy and estimate prognosis.
198
CANCERS OF THE FEMALE GENITAL SYSTEM 13
A B
Figure 13.1
A&B, CT scan in ovarian cancer. Contrast enhanced CT in a patient with advanced ovarian cancer demonstrates
bilateral solid (black arrows) and cystic (white arrows) adnexal masses and omental cake (white arrow heads).
Courtesy of Dr. E Sala, University of Cambridge.
Early stage disease (stage I and II) improves survival. Patients with mucinous
Surgery tumours should have an appendicectomy, as this
Initial management in early stage disease is may be the site of the primary.
maximal cytoreductive surgery and surgical
staging. Around one-third of patients with appar- Fertility preserving surgery
ent stage I–IIA disease will have a higher stage In younger patients with stage IA tumours
of disease after full staging, mostly stage III and favourable histology, unilateral salpingo-
(77%). The frequency of lymph node involve- oophorectomy and staging may be carried out,
ment for apparent stage I disease is 20%. There although data on fertility preserving surgery is
is no evidence that total lymphadenectomy limited. Endometrial biopsy should be carried
199
II SITE-SPECIFIC CANCER MANAGEMENT
• CA-125
• Transvaginal and/
or abdominal U/S
Interval Palliative
debulking chemotherapy
surgery or best
supportive
care or trial
Figure 13.2
Management of ovarian cancer.
Neoadjuvant chemotherapy
A randomized trial (EORTC 55971) has shown Table 13.1: Prognosis of ovarian cancer
that neo-adjuvant chemotherapy has equivalent FIGO 5-year survival 5-year disease-free
efficacy with lower morbidity than primary stage (%) survival
debulking surgery in patients with stage bulky III/ I 80–90 70–85
IV disease. Neo-adjuvant chemotherapy usually II 65–80 55–65
comprises 3 cycles of carboplatin and paclitaxel IIIa 50 45
(or carboplatin alone) followed by interval
IIIb 40 25
debulking surgery for patients responding to
chemotherapy, then a further three cycles of adju- IIIc 30 20
vant carboplatin and paclitaxel. Approximately IV 15 10
10% of patients will progress on chemotherapy
(platinum refractory) and the prognosis for these
patients is poor and not improved by surgery. For
this group further treatment is with a change of
chemotherapy or best supportive care. Prognosis
Adjuvant chemotherapy Prognosis according to stage is shown in Table
Adjuvant chemotherapy is aimed at improving 13.1. The most important prognostic variables
overall survival. There are no studies comparing after stage are in order; degree of differentiation,
best supportive care with chemotherapy. Meta- cyst rupture, substage of disease and age.
analyses suggest that adjuvant treatment with
Relapse
platinum compared with non-platinum mono-
therapy achieves a 30% relative risk reduction in Despite 70% of patients with advanced disease
mortality implying some benefit from platinum having a complete clinical response at the end of
based chemotherapy. Four phase III trials have surgery and chemotherapy (no detectable disease
investigated the benefit of adding taxanes to on CT scan and/or normal CA-125), 70% of
platinum. Two trials (GOG111 and OV10) these will relapse, with a median progression free
showed a mean survival benefit of 10–14 months survival from diagnosis of 16 months. The time
whist other two (ICON3 and GOG132) did not. from first relapse to death is now around 2 years.
A meta-analysis of the four trials demonstrates a There are no studies comparing best supportive
small survival advantage with the addition of care with chemotherapy in relapsed patients,
paclitaxel to platinum. Many expert consider the but the recent studies suggest that chemotherapy
combination of carboplatin and paclitaxel as the improves survival.
standard adjuvant chemotherapy. Presentation of relapse
The choice between single agent carboplatin
55–70% of patients present with an asympto-
and carboplatin with paclitaxel as first line treat-
matic rise in CA-125. The median time from this
ment should be made after discussion of the risks
to onset of symptoms is 3 months. 30–45% of
and benefits with the patient. Since platinum
patients present with symptoms, of whom 30%
exerts the major effect, and paclitaxel increases
relapse in the abdomen, 36% in the pelvis, 14%
toxicity, single agent carboplatin will be the
in abdomen and pelvis, and the remainder have
treatment of choice for patients with poor per-
distant metastases (frequently liver metastases).
formance status or co-morbidities.
Relapse may be defined by conventional
Intraperitoneal (IP) chemotherapy RECIST criteria (p. 44) or by the gynaecological
A number of studies have investigated intra cancer inter-group (GCIG) CA-125 criteria.
peritoneal chemotherapy. Although some have CA-125 relapse for those who had elevated pre-
shown a small improvement in PFS and OS treatment value which normalized after first line
(GOG-172), the toxicities were high (p. 38). In treatment (60% of all new patients) is defined as
the UK, IP chemotherapy is not currently offered ≥2× upper limit of normal on two occasions not
outside a clinical trial. less than 1 week apart. Patients in whom CA-125
202
CANCERS OF THE FEMALE GENITAL SYSTEM 13
Figure 13.3
Relapsed epithelial ovarian cancer
Treatment of relapsed ovarian
cancer.
Platinum refractory/resistant Partially platinum-sensitive Platinum-sensitive
was elevated but never normalized (30% of all has been suggested that the response rate to plati-
new patients): relapse is defined as a CA-125 value num can be improved by ‘artificially’ extending
≥2× nadir value on two occasions. The definition the platinum free interval by using non-platinum
of relapse in patients with normal CA-125 prior agents, but this has not been proven.
to initial treatment (10% of all new patients) is
similar to that of patients with elevated CA-125 Platinum sensitive disease
which normalized after first line treatment. Patients with a PFI of >18 months show response
rates of 60–95% to second line therapy, with a
Treatment (Figure 13.3) median time to progression of 10–12 months.
Chemotherapy for relapsed disease is palliative Those with a PFI of 12–18 months show response
and aims to prevent or treat symptoms in order rates of 50–60%, and a time to progression of
to improve quality of life and possibly extend approximately 9 months. Patients with a PFI of
survival. In selected cases with a long disease 6–12 months show response rates of 25–35% to
free interval (exceeding 12 or 18 months) and platinum chemotherapy and a median time to
localized relapse, surgery may be an option and progression of <6 months.
long term disease free survival may be possible. ICON4 showed an improvement in response
The probability of response to re-treatment rate for carboplatin and paclitaxel compared
with platinum chemotherapy depends upon the with carboplatin alone (57% vs. 50%), progres-
interval between the completion of platinum- sion free survival (12 months vs. 9 months), and
based first line therapy to the time of relapse, 2-year survival (57% vs. 50%) and median sur-
known as the platinum free interval (PFI). A vival (29 vs. 24 months). In relapsed platinum
longer PFI is associated with a higher response sensitive disease, combination treatment is there-
rate, longer PFS and improved survival. fore recommended unless there is significant
Disease relapsing >6 months after platinum residual neuropathy. Gemcitabine and carbopla-
treatment is considered ‘platinum sensitive’, tin is sometimes used as combination therapy, as
however this group is subdivided into ‘relatively’ phase III trial results show similar improvements
platinum sensitive (PFI 6–12 months) and fully in response rates and progression free survival to
platinum sensitive (PFI >12 months). carboplatin and paclitaxel.
Although most of the data about the PFI and In patients with a PFI of 6–12 months, the
its relation to response is taken from first relapse benefits of combination chemotherapy are re
studies, this finding has been extended to second duced. Single agent carboplatin is more com-
and subsequent relapses. Standard practice is monly offered in this group. An alternative that
continue re-treating with platinum (or platinum may be considered in this group is peglyated
combination) until the patient becomes platinum doxorubicin (caelyx), since response rates from
resistant, regardless of the line of treatment. It non-comparative phase III trials are similar.
203
II SITE-SPECIFIC CANCER MANAGEMENT
E
E
A B
Figure 13.4
MRI in endometrial cancer. Sagittal T2-weighted (A) and sagittal dynamic contrast enhanced T1-weighted (B) images
in a patient with stage 1B endometrial carcinoma demonstrate a large endometrial tumour (E) which is invading the
outer myometrium (black arrows). The tumour is extending to the endocervix, but the cervical stroma (white arrows)
is intact. Courtesy of Dr. E Sala, University of Cambridge.
206
CANCERS OF THE FEMALE GENITAL SYSTEM 13
Stage II
Stage I
No further EBRT +/– BT BT for EBRT +/– No further EBRT +/– BT EBRT +/– BT
adjuvant higher risk cases BT treatment
treatment follow
up in unit
Stage III and IV
Hysterectomy and BSO EBRT and BT then Stage III locally • Palliative hormones
and cytoreduction reassess advanced disease • Palliative chemotherapy
and lymphadenectomy • Palliative RT for symptom
(if involved) control
• Palliative surgery for symptom
control e.g. hysterectomy for
Optimal debulking bleeding pain
Adjuvant RT Adjuvant
chemotherapy if fit
Figure 13.5
Management of endometrial cancer.
208
CANCERS OF THE FEMALE GENITAL SYSTEM 13
Risk of locoregional
relapse
Serous papillary or High
Low <5%
clear cell
Intermediate 5–15%
High >15%
hysterectomy with bilateral lymph node dissec- retrospective studies suggest that optimal tumour
tion is indicated. This involves resection of a cytoreduction may improve survival. The defini-
parametrial and paracervical tissue, and a cuff of tion of optimal cytoreduction varies in different
2 cm of upper vagina. Evidence show that radical studies from no macroscopic disease to <2 cm.
hysterectomy results in an improved survival in One study found a 5-year survival of 41% in
stage II patients compared to standard hyster patients with stage III disease had a complete
ectomy (93% vs. 89% at 5 years). For patients resection of macroscopic tumour compared with
with adequate tumour free margins and negative 16% for those without. In cases of suspicious
nodes there is no evidence that the addition of operability, a diagnostic laparoscopy should be
radiotherapy improved survival. Those with pos- carried out. In patients with borderline operabil-
itive margins or involved nodes should receive ity, primary radiotherapy may be used to improve
adjuvant EBRT and BT. chances of subsequent surgery.
For some patients, endocervical stromal Most stage IV tumours will not be considered
involvement (stage II) will have been an inciden- operable. However, in a subgroup of stage IV
tal finding following simple hysterectomy for patients where complete resection is feasible, a
presumed stage I. These patients would normally 25% long-term survival can be achieved.
receive adjuvant EBRT and BT. In practice, many Palliative hysterectomy is useful in controlling
patients are not fit enough for modified radical local symptoms such as bleeding or pain. Occa-
hysterectomy and these patients are treated with sionally bowel or urinary diversion may be
standard hysterectomy with adjuvant EBRT appropriate, although radiotherapy and/or plati-
and BT. There is no proven role for adjuvant num based chemotherapy may also offer useful
chemotherapy for stage II disease yet. palliation.
Stage III–IV Radiotherapy (Box 13.7)
The optimal management of patients with Postoperative EBRT and BT should be carried
advanced stage endometrial cancer is not well out for patients with a complete macroscopic
defined. Advanced stage endometrial cancers are resection of stage III disease. Patients with stage
a heterogeneous group and treatment needs to be IIIC disease involving the para-aortic or pelvic
individualized. nodes may have extended field radiotherapy
covering the para-aortic region. The likelihood
Surgery of local control is related to disease volume.
Primary surgery should be considered for all fit Although radiotherapy reduces the local recur-
patients with stage III disease in whom an optimal rence rates, there are no data showing improved
cytoreduction appears to be feasible. Several survival.
210
CANCERS OF THE FEMALE GENITAL SYSTEM 13
stabilization rate of 39% for a median of 6.7 that surgery alone is inadequate. Hence, all
months with 2.5 mg letrozole in patients with women with resected stage IB, IC, and II USPC
metastatic or recurrent endometrial cancer. or clear cell cancers may be offered platinum
based chemotherapy (usually with carboplatin
Recurrent disease
and taxol) with postoperative radiotherapy
Approximately two-thirds of relapses will either (EBRT+VBT, VBT or EBRT alone) in those with
be distant, or locoregional and distant. However a complete clinical response. Most centres would
the other third will be a pelvic recurrence, of also offer adjuvant chemotherapy and radiother-
which 75% will be the upper vagina, or ‘vaginal apy for stage IA serous papillary carcinoma in
vault’. which there was any residual disease at hysterec-
For patients with a vault recurrence who have tomy following biopsy. Some centres consider
not previously received radiotherapy, radical radi- whole abdominal radiation for UPSC because of
otherapy is the treatment of choice (Box 13.8). the high rate of abdominal relapse, though the
Those patients who had previously received only benefit has not been established.
BT can have EBRT with a boost if tumour is Surgery and chemotherapy for stage III and IV
<0.5 cm. In the PORTEC-1 study 75% of patients UPSC and clear cell carcinoma is as for ovarian
with pelvic recurrence could be treated with cura- cancer. Radiotherapy is offered to those who
tive intent and 85% achieved complete remission. have had a complete clinical response.
The survival rate after relapse was 69% at 3 years
in the control group compared with 13% in Synchronous cancers
patients who had received EBRT up-front. Synchronous primary cancers of the ovary are
In single recurrence at the vaginal vault, or found in 5% of patients with endometrial cancer.
localized pelvic disease not extending to the side- Management is the combined treatment for each
wall after previous pelvic radiotherapy, surgery separate cancer. Metastatic disease should be
may offer a chance of cure. If the bladder and considered where the disease on the ovary is
bowels are involved, then a large exenterative small volume, surface deposits, bilateral with
procedure may be required for complete resec- multinodular implants and with lymphovascular
tion. The surgical morbidity of exenteration is invasion of the ovarian cortex.
significant and is suitable only for selected
patients, after metastatic disease has been care- Follow-up
fully ruled out. Reported 5-year survival is up to There is no evidence for best follow-up. The
of 50% with a complete cytoreduction. majority of recurrences (80%) occur in the first
Patients not suitable for salvage surgery or 18 months to two years. A typical follow up
radiotherapy may be treated with systemic schedule would be outpatient appointments
therapy. three-monthly for two years, followed by six-
monthly to 5-years with full history and physical
Special situations examination (including pelvic) examination.
Serous papillary and clear cell carcinoma
Patterns of spread for uterine serous papillary Cervical cancer
carcinoma (USPC) are similar to epithelial
ovarian carcinoma and 50% of recurrences occur Introduction and aetiology
in the abdomen alone. Clear cell carcinoma Cervical cancer affects 520,000 women each year
relapses occur more commonly in the pelvis and worldwide. In the UK around 2900 women are
para-aortic nodes and less commonly in the diagnosed with cervical cancer per year which
abdomen than UPSC. represents 2% of female cancer. It is the second
The principles of surgery for both these most common cancer in women under 35 (after
tumours parallel those for ovarian cancer (p. breast cancer). In the UK, the life time risk of
198). Optimal therapy of early-stage papillary developing risk of cervical cancer is 1 in 116.
serous and clear cell carcinomas remains unde- There are a number of risk factors implicated
fined. However the high relapse rates indicate in the development of cervical cancer such as
212
CANCERS OF THE FEMALE GENITAL SYSTEM 13
sexual activity under the age of 20, smoking, oedema, and hydronephrosis is associated with
immunosuppression, and infection with sexually extensive pelvic wall involvement. Patients with
transmitted diseases. Human papilloma virus advanced disease may present with haematuria or
(HPV) has emerged as the principal causative symptoms of a vesicovaginal fistula. Cachexia,
agent in the majority of cases of cervical cancer. cough, jaundice, and a left supraclavicular nodal
The most frequent subtypes are 16 or 18. mass may occur with distant metastases.
Pathogenesis and pathology Evaluation and staging
Most cervical carcinomas arise at the squamo- Pretreatment evaluation
columnar junction of the ectocervix and endocer- • Examination under anaesthesia (EUA).
vix. There is a progression from dysplastic lesions • FBC, and biochemistry to exclude anaemia
to invasive carcinoma. Dysplastic lesions undergo and renal impairment.
spontaneous regression in 25–38%, persist in • Biopsy for histological diagnosis.
50–60% and progress to invasive cancers in • Chest X-ray.
2–14%. • MRI pelvis (Figure 13.6) is useful in assessing
Squamous cell and adenocarcinomas account tumour extent and nodal status.
for 90–95% of the cervical cancers. Other histo- • Cystoscopy/sigmoidoscopy/barium enema/
logic types are shown in Box 13.10. IVU – if there is clinical suspicion of bladder,
Patterns of spread rectal or ureteric involvement.
• PET/CT has a role in accurate selection of
From the cervix, the tumour may extend locally patients for surgery as well as for treatment
to the lower uterine segment, vagina, or into the planning.
paracervical spaces. It may become fixed to the • CT scan chest, abdomen and pelvis are indi-
pelvic wall by direct extension or due to regional cated in patient with clinically apparent stage
adenopathy. IV disease.
Cervical cancer follows a pattern of metastatic
progression, initially to nodes in the pelvis and Staging
then para aortic nodes and distant sites. The The FIGO staging is given in Table 13.5 which
most frequent sites of distant recurrence are lung, does not take into consideration the findings on
extra pelvic nodes, liver, and bone. imaging.
Presentation Management (Figure 13.7)
Early invasive disease may be asymptomatic. The Choice of treatment is based on tumour size,
earliest symptom of invasive cervical cancer is stage, histology, evidence of lymph node involve-
usually abnormal vaginal bleeding, often post- ment, risk factors for complications of surgery or
coital. Pelvic pain may result from loco-regionally radiotherapy, and patient preference.
invasive disease. The triad of sciatic pain, leg In situ disease (CIN III) can be treated by a
number of methods (such as cone biopsy, laser
therapy etc.) depending on the extent of disease,
age of the patient, and requirement for fertility
Box 13.10: Histopathologic subtypes of preservation.
cervical cancer
• Squamous carcinoma: keratinizing, non-
Stage IA disease
keratinizing and verrucous Stage IA1
• Endometrioid adenocarcinoma IA1 without lymphovascular invasion (LVI) the
• Clear cell adenocarcinoma treatment options are:
• Adenosquamous carcinoma
• Adenoid cystic carcinoma • Cone biopsy or large loop excision of the
• Small cell carcinoma transformation zone (LLETZ) if patient
• Undifferentiated carcinoma wishes to preserve fertility.
• Simple hysterectomy.
213
II SITE-SPECIFIC CANCER MANAGEMENT
A B
Figure 13.6
MRI scan in cervical cancer. Sagittal (A)
and axial (B&C) T2-weighed MRI in a
patient with stage 2B squamous cervical
cancer demonstrate a large cervical
tumour (C) with bilateral parametrial
invasion (black arrows) and bilateral
enlarged external iliac lymph nodes (white
arrows). Note the presence of cystic/
necrotic changes within the right external
iliac node (white arrow head) which is a
common feature of squamous cell cervical
C cancer. Courtesy of Dr. E Sala, University of
Cambridge.
Cervical cancer
Cone biopsy
PA node + PA node –
Margin –, ≤3 mm Margin + or >3 mm Radical RT or radical Radical hysterectomy Extended field Chemo-RT
depth of invasion depth of invasion chemoradiotherapy and pelvic node dissection RT and
and no LVI or LVI chemotherapy
Recurrence
Figure 13.7
Management of cervical cancer.
217
II SITE-SPECIFIC CANCER MANAGEMENT
Figure 13.8
A,B – Treatment volumes
for external beam
radiotherapy in cervical
cancer C,D – Brachytherapy
dose distribution and
dosimetric points (AR –
right side point A, AL – left
side point A, BR – right side
point B, BL – left side point
B, ICRU bladder – bladder
point and ICRU rectum –
rectal point).
A B
C D
The majority of stage IVA patients are of poor Recurrence after primary surgery
performance status or with many co-morbidities Relapse in the pelvis following primary surgery
and extensive local disease, and are best treated may be treated by radiotherapy or pelvic exenter-
with palliative treatment (radiotherapy or chem- ation. Radical radiotherapy (+/− concurrent
otherapy). Patients with poor general health, or chemotherapy) may cure a substantial propor-
extensive local disease can be offered best sup- tion of those with isolated pelvic failure after
portive care. primary surgery. Radiation dose and volume
Stage IVB should be tailored to the extent of disease. Where
No standard chemotherapy regimen is proven in disease is metastatic or recurrent in the pelvis
patients with stage IVB cervical cancer. Radia- after failure of primary therapy and not curable,
tion therapy can be used for palliation of central a trial of cisplatin chemotherapy with palliative
disease or symptomatic distant metastasis. The intent or symptomatic care may be considered.
role of systemic therapy is discussed later. The expected median time to progression or
death is three to seven months.
Recurrence
Treatment decisions should be based on the Recurrence after primary radiotherapy
performance status of the patient, the site and Selected patients may be considered for pelvic
extent of recurrence and/or metastases, and prior exenteration, which is the only potentially cura-
treatment. tive treatment after primary irradiation. Those
218
CANCERS OF THE FEMALE GENITAL SYSTEM 13
with resectable central recurrences that involve Treatment toxicities
the bladder and/or rectum without evidence of Significant surgical complications are uretero-
intraperitoneal or extra pelvic spread and who vaginal fistula (<2%) and vesicovaginal fistula
have a dissectable tumour-free plane along the (<1%). The risk of complications increases with
pelvic sidewall are potentially suitable. Surgery addition of radiotherapy. Extensive pelvic fibro-
should be undertaken only in centres with appro- sis can lead to ureteric obstruction and small
priate facilities and expertise. The prognosis is bowel obstruction.
better for patients with a disease-free interval The late sequelae following radiation therapy
greater than six months, a recurrence 3 cm or commonly affect the rectum, bladder and small
less in diameter, and no sidewall fixation. The bowel. These depend on the duration of follow-
5-year survival for patients selected for treatment up, type of treatment modalities and estimated
with pelvic exenteration is between 30–60% and radiation doses to these organs. The reported
the operative mortality should be <10%. grade III/IV late sequelae range from 5–15%.
Role of chemotherapy in distant Palliative care
metastasis and recurrent
Palliative care issues include pelvic pain, bleed-
metastatic disease ing, bone pain from metastases and systemic
Chemotherapy has a palliative role after failure symptoms for advanced cancer.
of surgery or radiotherapy. There are a number
of agents with activity in metastatic or recurrent Follow-up
cervical cancer. Cisplatin is the most active agent, Optimal follow-up strategy has not been estab-
with a response rate of 20–30% and a median lished. A general guideline is for clinical evalua-
survival of 7 months. Recent studies suggest that tion three monthly for one year, four-monthly
cisplatin in combination with paclitaxel or topo- for one year, 6-monthly for three years and then
tecan is superior than cisplatin alone. annually. Patients are advised to have annual
chest X-ray. Other investigations are dictated by
Prognosis clinical indications.
Stage is the single most important prognostic
factor for local control and overall survival. Sur- Screening
vival rate after radical hysterectomy for stage IB The main goal of screening is to reduce the inci-
disease, is around 85–95% for node negative and dence and mortality. Screening has been shown
45–55% for node positive disease. Survival rates to be an effective method of identifying pre-neo-
for patients with para-aortic nodal disease treated plastic disease and reducing mortality. Cervical
with extended-field radiotherapy vary between Cytology (Pap Smear) Screening programs have
10% and 50% depending on the extent of pelvic been found to be successful in reducing cervical
disease and para-aortic lymph node involvement. cancer incidence. In the UK, women aged 25
Other factors associated with a poor prognosis years receive the first invitation for screening,
are LVSI, deep stromal invasion (10 mm or those aged 25–49 years have 3 yearly screening,
more, or more than 70% invasion) and parame- those aged 50–64 years have 5-yearly and those
trial extension. Stage-wise 5-year survival is as above 65 years have screening test only if they
follows: have not had screening since 50 years or have
• Stage I A – 95–97%. had a recent abnormal test.
• Stage I B1 – 89%.
• Stage IB2 – 76%. Prevention
• Stage IIA – 73%. HPV vaccines have been shown to reduce the
• Stage IIB – 66%. number of further pre-invasive lesions and inva-
• Stage III – 40%. sive disease in young women with pre-invasive
• Stage IVA – 22%. lesions. In the UK, there is a national programme
• Stage IVB – 9%. to vaccinate girls aged 12–13 against HPV.
219
II SITE-SPECIFIC CANCER MANAGEMENT
Postoperative management
Dysgerminoma
Adequately staged FIGO IA dysgerminoma has
long-term survival of more than 90%, and these
Figure 13.9 patients are observed without any postoperative
Dysgerminoma of the ovary. Contrast-enhanced CT scan
shows a large, multilobulated solid mass with highly
treatment. The recurrences (rate of 15–25%) in
enhancing fibrovascular septa (arrows) and cystic change these patients can be effectively salvaged with
(arrowheads). chemotherapy. All other patients need adjuvant 221
II SITE-SPECIFIC CANCER MANAGEMENT
Non-dysgerminoma Dysgerminoma
Consider VeIP or TIP Follow-up VeIP or TIP VeIP or TIP VeIP or TIP
resection
Box 13.13: Indications for chemotherapy in Box 13.14: FIGO staging of GTD
persistent trophoblastic disease
• Stage I – Disease confined to uterus
• Serum hCG >20,000 iu/l after one or two uterine • Stage II – disease beyond uterus but confined to
evacuations genital structures
• Static or rising hCG levels after one or two uterine • Stage III – lung metastasis with or without genital
evacuations tract involvement
• Persistent hCG elevation six months post-uterine • Stage IV – distant metastasis other than lung
evacuation
• Persistent vaginal bleeding with raised hCG levels
• Pulmonary metastasis with static or rising hCG
levels
• Metastasis in liver, brain, or GI tract Box 13.15: Chemotherapy in GTD
• Histological diagnosis of choriocarcinoma Low risk
• Methotrexate with folinic acid
• D-actinomycin
Salvage chemotherapy for low risk and first line
Assessment prior to chemotherapy chemotherapy in high risk
• MEA (methotrexate, etoposide and dactinomycin)
Investigations • EMA/CO (methotrexate, etoposide, dactinomycin/
Patients need further investigations with FBC, cyclophosphamide and vincristine)
biochemistry, serum beta hCG, chest X-ray and Salvage chemotherapy for high risk
CT scan of thorax and CNS staging in high risk • EMA/EP (methotrexate, etoposide, dactinomycin/
etoposide, cisplatin)
patients (WHO score of ≥7, high risk score, mul-
tiple lung metastases and hCG >50,000 iu/l).
CNS staging consists of imaging the cranio
spinal axis and CSF examination (ratio of Treatment
CSF: serum hCG > greater than 1 in 60 suggests
Low-risk disease (score ≤6)
involvement).
Single agent chemotherapy is the treatment
Staging and prognostic scoring of choice and the commonly used agents are
FIGO staging (Box 13.14) is mainly used to methotrexate and dactinomycin (Box 13.15).
compare outcome data and the scoring system Methotrexate does not cause alopecia. Around
(Table 13.6) guides treatment. Low risk disease 10–20% patients do not respond to methotrex-
is defined by a total score of ≤6 and high risk ate, and salvage treatment options are single
score ≥7. agent D-actinomycin (if hCG level is low) or
225
II SITE-SPECIFIC CANCER MANAGEMENT
combination chemotherapy (if hCG level is high) given. There is no consensus on when to stop
(Box 13.15). Selected patients are salvaged with chemotherapy after biochemical remission. Many
hysterectomy. patients receive 1–3 cycles of chemotherapy after
biochemical remission, depending on the risk
High risk disease (score ≥7)
category and the rate of reduction of hCG.
Patients with high risk disease are treated with
combination chemotherapy with or without Follow-up and survivorship
surgery. The commonly used combination chem-
otherapy regimes are shown in Box 13.15. Most Patients need regular follow-up after completion
of the chemotherapy regimes give an 80–85% of treatment. Women are advised not to conceive
complete remission. for at least 12 months after completion of their
treatment. Women are advised not to take the
Relapse or resistant disease oral contraceptive pill until hCG is normal.
20–30% of patients will be resistant to treatment There is no evidence that treatment for a
or relapse after initial treatment. The commonly previous GTD affects future pregnancy and its
used regime is EMA/EP (Box 13.15). There is a outcome. Chemotherapy generally results in an
role for surgical salvage in selected patients. early menopause such that patients are advised
to complete their families before the age of 35.
Central nervous system disease
There is also a risk of second tumours such as
Patients with CNS disease are treated with chem- AML.
otherapy with or without surgery or radiother-
apy. Modifying EMA/CO chemotherapy with an
increased dose of systemic methotrexate and Vulval cancer
with the addition of intrathecal methotrexate
may improve the dose of methotrexate in CNS. Introduction
Vulval cancer is rare, with 1022 new cases being
Placental site trophoblastic diagnosed in the UK each year. It predominantly
tumour (PSTT) affects older women, with 80% of cases occurring
PSTT is less chemosensitive than other GTDs in those over 60. HPV (mainly 16, 18 and 31) has
and hence hysterectomy is the treatment of choice been found to be responsible for approximately
for disease limited to uterus. Metastatic PSTT is 30–50% of vulval cancers and up to 80% of
treated like high risk GTD. vulval intraepithelial neoplasias (VIN). There is
also an increased risk in those with HIV as well
Assessing response to treatment and as those on immunosuppressions. Other risk
number of courses of chemotherapy factors include smoking and chronic skin condi-
In patients receiving chemotherapy for low-risk tions such as lichen sclerosus (a 4–7% risk of
disease, patients are closely monitored after one malignancy), lichen planus and Paget’s disease.
course of chemotherapy with weekly hCG.
Further treatment is withheld as long as serum Pathology
hCG is falling. A second course of treatment is There are two varieties of intraepithelial neopla-
indicated if the hCG levels are stationary for sia: squamous cell carcinoma-in-situ (Bowen’s
three consecutive weeks, re-elevates or does not disease) or vulvar intraepithelial neoplasia III and
decline by one logarithmic fall within 18 days of Paget’s disease.
completion of the first treatment. If a second The majority (90%) of vulval cancers are
course of chemotherapy is necessary, the same squamous cell (SCC) in origin, however, melano-
treatment is given if there was an adequate mas (4%), basal cell carcinomas, adenocarci
response to previous treatment. An adequate nomas (1–2%), undifferentiated carcinomas,
response is defined as at least a logarithmic fall sarcomas (<2%) and metastatic tumours from a
in hCG after a course of chemotherapy. If the variety of primary sites also occur. Verrucous
response is not adequate, another single agent SCC carcinomas are a slow growing type of
chemotherapy or combination chemotherapy is squamous cell carcinoma.
226
CANCERS OF THE FEMALE GENITAL SYSTEM 13
absence of nodal metastases. Stage III – Tumour of any size with or without
extension to adjacent perineal structures ( 13 lower
urethra, 13 lower vagina, anus) with positive
Clinical features inguino-femoral lymph nodes.
• IIIA
The most common symptoms include itch or irri-
(i) With 1 lymph node metastasis (≥5 mm), or
tation, pain and soreness, a thickened, raised
(ii) 1–2 lymph node metastasis(es) (<5 mm)
area of discolouration, ulcer, vaginal discharge
• IIIB
or bleeding, or lump.
(i) With 2 or more lymph node metastases
(≥5 mm), or
Diagnosis and staging (ii) 3 or more lymph node metastases (>5 mm)
The size and location of the lesion should be • IIIC With positive nodes with extracapsular
spread
documented, and any involvement of adjacent
Stage IV – Tumour invades other regional ( 2 3 upper
structures such as vagina, urethra, base of bladder urethra, 2 3 upper vagina), or distant structures.
or anus, should be noted. The diagnosis of vulval • IVA Tumour invades any of the following:
cancer is made after biopsy. Vulval cancer is (i) upper urethral and/or vaginal mucosa,
staged using the FIGO classification (Box 13.16). bladder mucosa, rectal mucosa, or fixed to
pelvic bone, or
(ii) fixed or ulcerated inguino-femoral lymph
Management (Figure 13.11) nodes
Early-stage disease (Stages I and II) • IVB – Any distant metastasis including pelvic
lymph nodes
Surgery is the treatment of choice which involves
a wide and deep resection (radical local excision)
to obtain surgical margins of at least 1 cm. All
patients except stage Ia (depth of penetration
<1 mm when the risk of node metastasis is <1%) Advanced disease
need an ipsilateral inguinofemoral lymphadenec- If it is possible to resect the primary lesion with
tomy to reduce the risk of recurrence. Bilateral clear margins and without the need for a bowel
lymphadenectomy is indicated if the tumour is or urinary stoma, primary surgery is desirable.
within 1 cm of midline or involving labia minor. Surgery involves radical vulvectomy, bilateral
Postoperative bilateral pelvic and groin irradia- groin node dissection and pelvic exenteration, or
tion is indicated in the following setting: ano-vulvectomy with formation of end colos-
tomy. The overall 5-year survival rate in women
• One macrometastases (>5 mm in diameter).
with locally advanced disease treated by radical
• Two or more micrometastases (≤5 mm).
ano-vulvectomy has been reported to be 62%.
• Extracapsular spread.
Postoperative radiotherapy is indicated if the
Radiotherapy field should include inguino- margin is <5 mm and re-excision is not feasible.
femoral nodes and at least pelvic nodes below Primary radiotherapy is the treatment of
the SI joints. Microscopic disease needs 50 Gy in choice in those women with larger, more
1.8–2 Gy per fraction, 60 Gy for extracapsular advanced lesions involving bladder or rectum, or
spread or multiple nodes and 60–70 Gy for who are considered unsuitable for surgery. Nodal
macroscopic disease. disease may be dissected prior to management of
227
II SITE-SPECIFIC CANCER MANAGEMENT
Histologically
confirmed vulval cancer
primary tumour. Initial treatment involves 50 Gy frequency, and the formation of fistulae (recto-
at 1.7–1.8 Gy per fraction to include pelvis, vaginal, vesico-vaginal and entero-vaginal). The
inguinal nodes and primary tumour. The second incidence of developing lower limb lymphoedema
phase of treatment is to the gross disease to is also greater in those women who have been
deliver a total dose of 60–70 Gy. The exact role treated by surgery including lymphadenectomy,
of the combination of chemotherapy (cisplatin and adjuvant radiotherapy (groin and pelvis).
alone or in combination with 5-FU) with radio-
therapy is not fully explored. Recurrence and management
15 and 33% of vulval cancers recur and the
Prognosis most common sites of recurrence are the vulva
Prognosis depends on nodal status and the size (69.5%), groin nodes (24.3%), the pelvis in
of the primary lesion. In the absence of lymph 15.6% and distant metastases in 18.5%.
node involvement, the 5-year survival is >80%. Local vulval recurrences are treated by surgery
This decreases to approximately 50% with when possible. Groin recurrences are more dif-
inguinal node involvement, and to 10–15% with ficult to manage. Radiotherapy is the preferred
pelvic node metastases. treatment, but surgery should be considered in
patients who have already received groin
Post-treatment issues irradiation.
Lymphoedema is a late complication, with an
incidence of 62–69% following groin node Cancer of the vagina
dissection. When it occurs, the onset of lym-
phoedema is apparent within three months in Introduction
50% of women, and within 12 months in 85% Primary vaginal cancer constitutes 2% of female
of women. genital tract cancers, and 240 new cases are diag-
Complications following radiotherapy include nosed annually in the UK. It causes approxi-
vulval soreness, skin blistering, diarrhoea, urinary mately 100 deaths in the UK each year.
228
CANCERS OF THE FEMALE GENITAL SYSTEM 13
extension outside the vaginal wall, surgery may vagina, and poorly differentiated tumours, are
be considered. poor prognostic factors.
Pelvic exenteration is useful in stage IVa disease
without extension of the lesion to the pelvic side Recurrence and management
walls, and distant sites of metastases. The main site of relapse is the pelvis. In central
In young patients requiring radiotherapy, recurrence, there may be a role for pelvic exen
laparotomy is done to transpose ovaries. teration or further radiotherapy. The 5-year
survival rate following recurrence is approxi-
Radiotherapy mately 12%. There is no proven benefit for
Radiotherapy is the treatment of choice unless chemotherapy.
clear resection margins (at least 1 cm) can be Follow-up
obtained. The proximity of the vagina to the
bladder and rectum can potentially limit treat- Follow-up is important in addressing the issues
ment options and increase the risk of complica- of treatment-induced morbidities. Vaginal scar-
tions to these organs. ring and stenosis are the main problems leading
In early stage disease (stage I and II), intra to depression and sexual dysfunction. Late
cavitary radiation may be used, while both intra bladder and bowel toxicities and early meno-
cavitary and external irradiation are required for pause due to radiation also need attention.
larger and more advanced lesions. The use Further reading
of both external irradiation and brachytherapy
in treating vaginal cancer has been shown to Colombo N, Peiretti M, Castiglione M. Non-epithelial
ovarian cancer: ESMO clinical recommendations for
achieve excellent results, and allows vaginal diagnosis, treatment and follow-up. Ann Oncol. 2009;20
preservation. If the lower one-third of the vagina Suppl 4:24–26.
is involved groin nodes should be irradiated or Han LY, Kipps E, Kaye SB. Current treatment and clinical
dissected. trials in ovarian cancer. Expert Opin Investig Drugs.
Women with advanced disease tend to be 2010;19:521–534.
Amant F, Moerman P, Neven P et al. Endometrial cancer.
treated with chemoradiation rather than radio- Lancet. 2005;366:491–505.
therapy alone. Gehrig PA, Bae-Jump VL. Promising novel therapies for the
treatment of endometrial cancer. Gynecol Oncol.
Chemoradiotherapy 2010;116:187–194.
Goonatillake S, Khong R, Hoskin P. Chemoradiation in
Most of the evidence for chemoradiotherapy in gynaecological cancer. Clin Oncol. 2009;21:566–572.
vaginal cancer originates from studies in cervical Taylor A, Powell ME. Conformal and intensity-modulated
cancer using cisplatin or cisplatin with 5-FU. radiotherapy for cervical cancer. Clin Oncol. 2008;20:
Chemotherapy alone appears to offer little benefit 417–425.
in the management of advanced (stage III and IV) del Campo JM, Prat A, Gil-Moreno A et al. Update on novel
therapeutic agents for cervical cancer. Gynecol Oncol.
disease. 2008;110:S72–S76.
Koulouris CR, Penson RT. Ovarian stromal and germ cell
Prognosis tumors. Semin Oncol. 2009;36:126–136.
The overall 5-year survival rate for vaginal cancer Crosbie EJ, Slade RJ, Ahmed AS. The management of vulval
cancer. Cancer Treat Rev. 2009;35:533–539.
is 44%, which is poorer than that for both cervi- Gray HJ. Advances in vulvar and vaginal cancer treatment.
cal and vulval cancer. Women over 60 years of Gynecol Oncol. 2010 May 13. [Epub ahead of print]
age, lesions in the middle and lower third of the PMID: 20471671
230
Cancers of the skin
HM Hatcher and TV Ajithkumar
14
Cutaneous melanoma • Naevi – Multiple benign naevi (>100) as well
as multiple aypical naevi increases the risk of
Epidemiology melanoma (RR 11)
Melanoma is the most aggressive form of skin • Immunosuppression – Transplant recipients
cancer which is increasing in incidence. In Europe, (RR 3) and patients with AIDS (RR 1.5) have
its incidence has risen by 3–8% per year since the increased risk of melanoma
1960s. The lifetime risk of melanoma in the UK • Previous melanoma – those with a previous
is 1 in 147 for men and 1 in 117 for women. In melanoma have a 2–10% risk of a further
Australia the risks are significantly higher with melanoma which increases further with two
lifetime risks of 1 in 25 for men and 1 in 35 for previous melanomas.
women. Overall survival rates have improved
due to early detection but survival of higher Pathology
stage disease has improved very little in the past There are four histological variants of cutaneous
10 years. melanoma:
The peak incidence of melanoma occurs in
middle age (40s) with less than 10% occurring • Superficial spreading is the most common
in childhood, although the rates in those under type. This often arises within a pre-existing
20 have increased by over 3% in the past naevus and is surrounded by a zone of atypi-
10 years. cal melanocytes that may extend beyond the
visible border of the lesion.
Aetiology • Nodular melanoma (10–15%) presents as
symmetrical, uniform dark blue-black lesions.
Ultraviolent sun radiation (especially UVB) is
Amelanotic nodular melanomas are often
the main risk factor for the development of
misdiagnosed.
cutaneous melanoma. People with a history of
• Lentigo maligna melanoma (10–15%) usually
blistering sunburns have a 2.5 times higher risk
occurs on the sun-exposed areas of head and
of melanoma. Other surrogate makers of sun
neck and hands. Clinically these are large
sensitivity such as freckling (RR 2.5), burn
(often >3 cm) flat lesions with areas of dark
without tanning (RR 1.7), red hair (RR 2.4) and
brown or black discolouration. These lesions
blue eyes (RR 1.6) also have an increased risk.
arise from the premalignant lesion called
Other risk factors are:
Hutchinson’s freckle.
• Genetic – people with a strong family history • Acral-lentigenous melanomas, as the name
and multiple atypical moles are at the suggests (acral – distal), occur on the palms,
greatest risk of melanoma. Those with an soles and subungual regions. These lesions
inherited mutation in the CDKN2A and occur with the same frequency in whites and
CDK4 genes have 60–90% lifetime risk of non-whites (2–8% of melanoma in whites
melanoma. and 40–60% melanoma of non-whites). These
Stage IB 91
T1bN0M0 ≤1 mm thickness with ulceration or mitosis ≥1/mm2
T2aN0M0 1.01–2 mm thickness without ulceration
Stage IIA 77–79
T2bN0M0 1.01–2 mm thickness with ulceration
T3aN0M0 2.01–4 mm thickness without ulceration
Stage IIB 63–67
T3bN0M0 2.01–4 mm thickness with ulceration
T4aN0M0 >4 mm thickness without ulceration
Stage IIC 45
T4bN0M0 >4 mm thickness with ulceration
Stage IIIA
T1-4aN1aM0 Micrometastasis* in 1 node 70
T1-4aN2aM0 Micrometastasis in 2–3 nodes 63
Stage IIIB
T1-4bN1a/2aM0 50–53
T1-4aN1bM0 Macrometastasis** in 1 node 46–59
T1-4aN2bM0 Macrometastasis in 2–3 nodes 46–59
T1-4aN2cM0 in-transit metastases/satellites without nodes
Stage IIIC 24–29
T1-4bN1b/2b/2cM0
Any T N3M0 metastasis in >3 nodes or matted nodes or in-transit or satellites with
metastatic nodes
Stage IV
Any T, any N, M1 distant metastasis 7–19%
*micrometastases after sentinel node biopsy; **macrometastases are clinically detectable pathologically confirmed nodes.
in patients with a high-risk resected melanoma. A trial examining the role of a ganglioside
This group included patients with ≥4 mm thick vaccine in the adjuvant treatment of high risk
with no nodal involvement and melanoma involv- melanoma showed no benefit and there were
ing regional nodes or in-transit metastasis. concerns that patients may have done worse than
However, high dose interferon is associated with with no treatment.
a number of side effects such as acute constitu-
tional symptoms, chronic fatigue, headache, Management of positive nodes
weight loss, nausea, myelosuppression and Patients with metastatic lymph node disease are
depression and most patients will require dose treated with regional node dissection if feasible
modifications during treatment. and up to 13–59% of these patients do not
234
CANCERS OF THE SKIN 14
develop metastatic disease. There is a significant been used to exclude further distant disease,
risk of lymphoedema (especially with groin dis- especially if complex surgery is planned, e.g.
sections) and patients should wear compression resection of cerebral metastases.
stockings for many months. Some trials are Patients with up to three visceral metastases are
examining the role of adjuvant treatment, such candidates for surgical resection and the 5-year
as bevacizumab, after groin dissection. A rand- survival can be more than 20% after complete
omized study has evaluated the role of postop- resection of lung metastases and 28–41% after
erative radiotherapy (48 Gy in 20 fractions) in complete resection of gastrointestinal metastases.
patients with a high risk (>25%) of regional Similarly patients with solitary brain metastases
recurrence after lymphadenectomy. The two- can also be candidates for resection. Adjuvant
year lymph node field relapse-free rate was sig- radiotherapy is often used in some of these patients,
nificantly improved with adjuvant radiotherapy especially after resection of cerebral metastasis.
compared with observation (82 vs. 65% HR
Role of radiotherapy
0.47, 95% CI 0.28–0.67); but without no
Radiotherapy can also be given after surgical
improvement in 2-year overall survival. The long
resection of a solitary metastasis and this is
term toxicity of radiotherapy yet to be reported.
particularly the case after resection of a solitary
brain metastasis. The treatment options include
Management of metastatic disease
whole brain radiotherapy (20 Gy in five fractions
Metastatic melanoma has a poor prognosis. Most or 30 Gy in 10 fractions), stereotactic radio
patients with non-visceral metastases survive up surgery or a combination of both. The optimal
to 18 months whereas the median survival of radiotherapy in this situation is yet to be defined.
those with visceral involvement or an elevated Palliative radiotherapy is useful in bone metas-
serum LDH is 4–6 months. Lymph node and skin tasis and spinal cord compression not amenable to
metastases in the absence of other metastases surgical decompression (p. 328). Skin metastases
have the best prognosis (up to 18 months). Those which are rapidly enlarging and about to ulcerate
with lung metastases in the absence of other vis- can also be treated with palliative radiotherapy.
ceral disease have an intermediate prognosis (up
to 12 months median survival). Those with other Chemotherapy and other systemic treatments
visceral disease have a median survival of less in metastatic disease
than 6 months which is limited further in the Dacarbazine is the standard intravenous chemo-
presence of a high and rapidly increasing LDH. therapy drug for metastatic melanoma with a
Patients with liver metastases from a primary response rate of 10–20% and a median duration
uveal melanoma have a particularly poor prog- of response of 3–6 months. It is given 3–4 weekly
nosis usually limited to less than 3 months. at doses of 850–1000 mg/m2 with nausea as the
Treatment of melanoma metastases depends main side effect. Temozolamide is an oral ana-
on the site of disease, whether or not it is local- logue of dacarbazine and crosses the blood–brain
ized and the overall fitness of the patient. Treat- barrier but is more expensive and has no improve-
ment can be with surgery, radiotherapy, systemic ment in response rates compared with dacar-
therapies or best supportive care. bazine (Table 14.2). There is no evidence that
combination chemotherapy regime is superior to
Role of surgery single agent drugs in terms of response rates or
In the presence of an isolated metastasis, espe- survival.
cially if the patient has a long disease free inter- Immunotherapy with high dose interleukin-2
val, e.g. 1 year, surgery may be the best option. (IL-2) or interferon alpha has shown response
The most common sites for surgical resection are rates of 10–21%. Toxicities include hypotension,
skin, brain and lung. Resection of liver metas- capillary leak syndrome, sepsis and renal failure.
tases is not usually performed as liver metastases A recent study of ipilimumab, a monoclonal
are associated with such a poor prognosis. antibody against cytotoxic T-lymphocyte antigen
Appropriate staging is important in these patients 4 (CTLA-4), showed improved overall survival
who undergo surgical resection. PET scans have in previously treated patients with unresectable
235
II SITE-SPECIFIC CANCER MANAGEMENT
Newer agents
Table 14.2: Response rates (%) for single Given the risk of recurrence and poor outcomes
agents in metastatic melanoma
with systemic treatment in metastatic disease,
% Response rate new agents are being developed to target specific
Agent (CR + PR)
pathways involved in melanoma. Examples
Dacarbazine 20
include VEGF, BRAF and bcl-2. Studies are
Temozolamide 21 ongoing to assess the potential benefit of bevaci-
Carmustine (BCNU) 18 zumab in resected stage III and stage IV disease
Lomustine (CCNU) 13
and the role of other anti angiogenics. Sorafenib
which is a TKI which has activity against BRAF
Cisplatin 23
has been shown to improve response rates when
Carboplatin 16 combined with chemotherapy, although no sur-
Vinblastine 13 vival benefit was observed. Agents with a more
Paclitaxel 18
specific BRAF activity are being developed.
Docetaxel 15
Prognostic factors and survival
Prognostic factors are highlighted in Box 14.1.
Survival according to stage is included in
stage III/IV melanoma who were positive for Table 14.1.
HLA-A*0201. Side effects of this treatment
included rash, colitis, diarrhoea and hepatitis. Follow-up
Combining chemotherapy with an immune
modulator has been assessed in several clinical After surgery all patients should be followed up
trials. A meta-analysis showed that this improved due to the risk of further melanomas as well as
response rates, but did not translate into a nodal or systemic recurrence. Full skin and nodal
survival benefit. examination should be carried out and abdomi-
In summary no drug or combination of drugs nal examination to exclude hepatomegaly. For
has shown a significant response rate and high-risk patients (>IA) blood tests to examine
survival benefit over single agent dacarbazine. It LDH, liver function tests and FBC may be carried
remains the standard drug in metastatic melanoma out. At the point of nodal recurrence reassess-
but patients should be entered into clinical trials ment with CT is suggested prior to surgery and
where possible. 6–12 months later. For those who have had met-
astatic disease resected a CT several months later
Isolated limb perfusion is recommended along with close follow-up. All
In some cases widespread metastases do not occur patients, especially who have had nodal disease
but there may be disseminated skin metastases in or metastases, should be warned about the symp-
a limb which cover an area too large for radio- toms of spinal cord compression.
therapy. In these patients, if systemic treatment is
not feasible, isolated limb perfusion with TNF-
alpha and melphalan can establish good local Non-cutaneous melanoma
control. This is only performed in a limited number Ocular melanoma
of centres but can provide good clinical benefit.
Ocular melanoma is a rare subtype of melanoma
Radiofrequency ablation which can occur in many areas of the eye. Most
For some patients with limited systemic disease commonly it occurs in the choroid but less than
but in whom surgery is not possible, radiofre- 5% occur in the iris. The aetiology is uncertain;
quency ablation (RFA) has been used for liver some studies suggest a link with UV radiation.
and lung metastases. In general they should be The risks are higher in those who have had cuta-
less than 5 cm in size and accessible to the RFA neous melanoma, pale iris colour or who have a
catheter used. Complications include bleeding family history of ocular melanoma. The staging
236 and pneumothorax. system differs to that of cutaneous melanoma.
CANCERS OF THE SKIN 14
Poor prognostic factors include age over 60, practice. A study using carbon ions to minimise
size and involvement of the ciliary body. Although local morbidity showed good local control but
often localized at presentation they have a 5-year survival was still less than 30%. Chemo-
high risk (10–80% depending on number of therapy with standard drugs for cutaneous
prognostic factors) to spread to the liver, often melanoma has been tried with similar success.
several years after successful treatment of the Nodal disease occurs to a greater extent than in
primary. Long-term survival is less than 35% ocular melanoma so regional lymph nodes should
even with successful treatment of the primary. always be examined at follow-up.
Surgery used to be the standard treatment but
radiotherapy has replaced this in most cases. Basal cell carcinoma
Radiotherapy can be administered as external
beam, as a radio-active plaque (e.g. iridium-192) Introduction
or with protons or other charged particles. Local Basal cell carcinoma (BCC) is a slow growing,
control rates are similar with each technique. The locally invasive (hence called rodent ulcer) malig-
vision is often lost in the irradiated eye with the nant epidermal skin tumour. The exact incidence
list of complications including cataracts, glau- is difficult to obtain; though there is a worldwide
coma, retinopathy and vitreous haemorrhage. trend in increasing incidence.
Treatment of metastases in ocular melanoma The most significant aetiological factors appear
has shown that they tend to have a reduced to UV exposure and genetic predisposition. BCC
response rate to chemotherapy and immuno- is common in the sun-exposed area of head and
therapy than cutaneous melanoma and are more neck. Multiple BCC is a feature of Gorlin’s syn-
rapidly progressive. drome. Other risk factors include increasing age,
Due to their propensity for metastases (espe- male, fair skin, immunosuppression, vaccination
cially liver and lung) which can be delayed, fol- scars and arsenic exposure. Sunscreen use and
low-up includes imaging of the liver and lungs in low fat diet are thought to be protective.
addition to LDH and liver function tests. Com-
bined follow-up with ophthalmology is required. Pathology
The common histologic subtypes are superficial,
Mucosal melanoma
nodular and morphoeic (sclerosing). Other vari-
Mucosal melanomas constitute less than 2% of ants include micronodular, infiltrative and baso
all melanomas. The most common sites of pres- squamous BCC which are aggressive with high
entation are head and neck (up to 50%), female local recurrence. Perivascular and perineural
genital tract (25%) and anorectal (20%). The invasion is associated with aggressive tumours.
remainder occur in very rare sites such as other BCC infiltrate tissues in a three dimensional
areas of the gastrointestinal tract, Eustachian fashion. Lymph node metastasis is extremely rare
tube and salivary glands. They occur later in life except in those with multiple recurrences or
(50–70s) than cutaneous melanoma, are more uncontrolled primary tumour.
common in non-Caucasians and slightly more
common in men than women. They tend to Clinical features
present late as they are essentially hidden from
The common growth patterns are superficial,
sight and have a very poor prognosis. For head
multifocal, nodular and morphoeic (Figure 14.2).
and neck tumours 5-year survival is less than
The sites affected are head and neck (52%), trunk
30% which reduces to <20% with involved
(27%), upper limb (13%) and lower limb (8%).
lymph nodes. The most common presentation is
with bleeding, anaemia or local symptoms such • Superficial BCC occurs on the trunk or limbs
as pain. There is no specific staging system. The of young people. It presents as a well-defined,
treatment of choice is surgical resection with clear erythematous, scaling or slightly shiny
margins but this is often difficult due to location macular lesion. Stretching the lesion causes an
of the tumour. Radiotherapy has been used to try increase in the degree of erythema, highlights
to improve local control but has not shown an the shiny surface and reveals a peripheral
improvement in survival and is not a standard thread-like pearly rim or island of pearliness 237
II SITE-SPECIFIC CANCER MANAGEMENT
Special considerations
Electron planning
• Electron beam field is defined by 50% isodose and the 90% isodose is 3–5 mm inside the field. Hence, in order
to enclose the PTV within 90% isodose, a 5 mm larger electron applicator than defined by PTV is needed.
• At higher energies, isodoses close to the surface bows inwards which necessitates 1 cm larger applicator
diameter than the defined PTV to ensure homogenous dose to the tumour (p. 18, Figure 3.2B).
• Surface dose increases with electron energy; a bolus is needed for lower energies to bring up 90% isodose to
the skin surface (p. 18, Figure 3.2A).
• Bolus is also used to bring up high dose to surface to avoid radiation to underlying critical structures.
• Stand off effect – fill the area with bolus/calculate correction
Normal tissue shielding
• Lower eyelid and canthi tumours need corneal shielding.
• Lip tumours need buccal shields. Shields should be coated with wax to absorb scattered radiation.
• Tumours in the pinna and nasal regions when treated with electrons need wax coated lead plugs in the
external auditory canal and nose respectively to avoid normal tissue damage.
240
CANCERS OF THE SKIN 14
metastasis, although progression to invasive SCC masses and bleeding. Metastasis is primarily to
occurs in 3–11% of cases. the regional nodes (2–6%). The head and neck
In situ SCC of the glans penis (erythroplasia region is the commonest site in men whereas the
of Queyrat) has 20% risk of metastasis and 30% upper limb followed by head and neck are the
can progress to invasive disease. commonest sites in females. Only 8% of SCC
Invasive SCC is composed of a collection of arise on the trunk.
atypical keratocytes which invade the dermis and
deeper structures. Other histologic variants are: Examination
Examination should be of the whole skin and
• Adenoid SCC – can metastasize in 3–19% regional lymph nodes.
and associated with rapid local growth.
• Adenosquamous SCC – shows squamous Diagnosis
appearance superficially and glandular appear- Tissue biopsy is essential for diagnosis. CT scan
ance deeply. These are aggressive tumours. helps to detect regional lymph nodes. FNA of the
• Spindle cell SCC – appears as ulcerated nodules enlarged lymph node under radiological guid-
or exophytic tumours and may be difficult to ance is advised. Open surgical biopsy should be
distinguish from sarcoma histologically. These avoided. The role of sentinel node biopsy is
tumours are aggressive with a tendency to evolving.
perineural invasion and metastasis (25%).
• Verrucous SCC – appears like a large wart. Staging
These are slow growing, locally invasive and T1 – ≤2 cm in greatest dimension.
do not metastasize. T2 – >2 to 5 cm in greatest dimension.
• Keratoacanthoma – appears as a rapidly T3 – >5 cm in greatest dimension.
growing nodule with a central keratin plug. T4 – tumour invades deep extradermal
True lesions can undergo spontaneous regres- structures.
sion but most are now viewed as SCC. N1 – regional nodal metastasis.
Clinical presentation M1 – distant metastasis.
Typical presentation is a raised pink papule or Treatment
plaque with erosion or ulceration (Figure 14.3). Three factors that influence treatment of SCC
In advanced cases it presents as large ulcerated are: the need for removal of tumour locally, the
possibility of ‘in-transit’ metastasis and regional
nodal metastasis. Treatment options include:
• Cryotherapy (for small tumours with well
defined borders).
• Curettage and electrodessication (for well
differentiated tumours of <1 cm size).
• Radiotherapy (see Box 14.3) – used as a
primary treatment and adjuvant after surgery.
• Surgical excision (see Box 14.3) and Mohs’
micrographic surgery.
Surgery
Surgery is aimed at complete removal of the
tumour and of any metastasis. Low-risk tumours
of <2 cm are excised with a minimal margin of
4 mm whereas tumours of >2 cm size, high risk
Figure 14.3
tumours (grade 2–4, in high risk locations such as
Digital squamous cell carcinoma. From Darrell Rigel: ear, lip, scalp, eyelids and nose) and those extend-
Cancer of the Skin (Saunders) with permission. ing into subcutaneous tissue need a minimum
241
II SITE-SPECIFIC CANCER MANAGEMENT
Keratoacanthoma
Presents as a rapidly enlarging lesion with central Figure 14.4
keratin plug. Histologically, it is difficult to Merkel cell carcinoma. From Darrell Rigel: Cancer of the
distinguish this from SCC. In up to 20% of Skin (Saunders) with permission.
patients spontaneous regression can occur over
6–12 weeks. Treatment options include early develop distant metastasis. Liver, lung, bone and
excision and radiotherapy (similar to SCC). brain are the sites of distant metastases.
Squamous cell carcinoma of lip Diagnosis and staging
Lesions involving <30% of lip can be treated Initial evaluation includes clinical examination
with wedge excision and repair. When >30% of of the whole skin surface and regional nodes.
the lip is involved, surgery results in functional Imaging includes CT scan of the relevant nodal
disability and hence radiotherapy is a treatment region as well as chest and liver. Blood tests
option. Treatment is with orthovoltage X-rays or include full blood count and biochemistry.
electrons with an intraoral lead shield to protect 70–80% patients present with local disease
the mandible or teeth or by implantation. Typical (stage IA ≤2 cm and IB >2 cm), 10–30% with
radiation dose is 50–55 Gy in 20–25 fractions. regional nodal disease (stage II) and 1–4% with
distant metastasis (stage III).
Merkel cell carcinoma
Treatment
Introduction
Stage I disease is usually treated with wide excision
Merkel cell carcinoma (MCC) is a rare aggressive of the primary tumour (2.5–3 cm margin) and
neuroendocrine tumour of the skin. The exact sentinel node biopsy. All patients receive postop-
aetiology is not known. The reported risk factors erative radiotherapy to the primary site with a
include ultraviolet exposure, previous skin generous margin of 3–5 cm (to ensure dermal
cancers and haematological malignancies, immu- lymphatics are treated). Regional lymph nodes are
nosuppression and HIV infection. The average generally treated electively due to the high risk of
age at presentation is 69 years. Males are more recurrence (46–76%) when nodes are not treated.
commonly affected. Radiotherapy dose ranges from 45–60 Gy using
Pathology 1.8–2 Gy per fraction. The role of adjuvant chem-
Histologically these are small blue cell neoplasms otherapy is unknown. Recent studies indicate that
which express neuroendocrine and cytokeratin primary radiotherapy (60 Gy) is an appropriate
markers. alternative which yields a better cosmesis.
Stage II disease is treated with surgery of the
Clinical features primary lesion and nodal dissection. All patients
MCC present as red or violaceous nodules with receive postoperative radiotherapy (50–60 Gy).
a shiny surface, often with overlying telangi Adjuvant chemotherapy may be considered.
ectasia (Figure 14.4). Spread through dermal Chemotherapy regime mirrors those used for
lymphatics can result in the development of satel- small cell lung cancer.
lite lesions. The most common site of disease is Stage III disease has a median survival of 9
the head and neck region, followed by extremi- months. Platinum based chemotherapy yields a
ties and trunk. One-third of patients have regional short lived complete response of 44% and partial
nodal metastases at presentation and 50% response of 11%. 243
II SITE-SPECIFIC CANCER MANAGEMENT
Staging
CT chest
Bone scan
Work up for treatment:
bloods, MUGA/ECHO, GFR
Localized Metastatic
Postoperative
chemotherapy
* Neoadjuvant chemotherapy PAM (cisplatin, doxorubicin and methotrexate) for young fit patients
and cisplatin/doxorubicin for older patients
combination chemotherapy should be the volume lung metastases, or with the intention of
primary treatment. cure, e.g. resection of lung metastases.
High-dose treatment with stem cell rescue has
Radiotherapy
shown no improvement in survival. Trials are
Although osteosarcomas are relatively radio
underway to evaluate biological agents such
resistant tumours, radiotherapy has been success
as insulin like growth factor 1 receptor (IGF1-R)
fully used in a number of clinical situations
antagonists. Immunotherapy may have a role
such as to control bone pain. It has also been
and the use of liposomal muramyl tripeptide-
used in addition to chemotherapy and surgery,
phosphatidyl-ethanolamine has been shown to
e.g. following resection of lung metastases some
produce a survival benefit in the adjuvant setting.
groups advocate whole lung radiotherapy in
Surgery addition to chemotherapy. It may provide local
Surgery in the metastatic setting should either be control for some patients with advanced meta
aimed at palliation of symptoms, e.g. resection static disease for whom resection of the primary
of a large primary extremity tumour with low is inappropriate.
249
II SITE-SPECIFIC CANCER MANAGEMENT
Biopsy
Staging
MRI of primary site
CT chest
Bone scan
Bone marrow
Work up for treatment:
bloods, MUGA/ECHO, GFR
Neoadjuvant
chemotherapy
e.g. 6 cycles of VIDE
Further chemotherapy
Figure 15.6 e.g. VAI, VAC
X-ray of Ewing’s sarcoma of right proximal femur with
destructive lesion and onion skin appearance of Figure 15.8
periosteal reaction. Courtesy of Mr Rob Grimer. Management of Ewing’s sarcomas (VIDE – vincristine,
ifosfamide, doxorubicin & etoposide; VAI – vincristine,
actinomycin-D & ifosfamide; VAC – vincristine,
actinomycin-D & cyclophosphamide).
Biopsy Surgery
Surgery of the primary lesion may still be appro
priate with limited pulmonary metastases if these
Staging respond to initial chemotherapy. Surgery may
MRI of primary site
also be necessary for local control, especially if
CT chest
Bone scan highly symptomatic.
Bone marrow
Work up for treatment: Radiotherapy
bloods, MUGA/ECHO, GFR Radiotherapy can be used to treat either the
primary tumour or metastatic lesions in advanced
disease. The dose (radical or palliative) used for
Neoadjuvant the primary will depend on the extent of other
chemotherapy
6 cycles of VIDE disease. Bilateral low-dose lung radiotherapy can
be used as an adjunct to chemotherapy for
patients with lung metastases who have had a
Surgery and/or radiotherapy good response to treatment. Unilateral lung irra
diation is used for symptomatic bulky chest wall
lesions. Palliative radiotherapy is used for painful
Assess response and consider
volume of disease bony metastases.
Prognosis of Ewing’s sarcoma
Good Poor Extensive disease Prognosis in localized disease has improved sig
nificantly since the introduction of chemother
apy. Poor prognostic features are listed in Box
VAI x 1 then VAI x 1 the High dose or
15.4. With combination chemotherapy and
VAC vs VAI x 7 VAI x 7 vs phase II trial
high dose surgery or radiotherapy 5-year survival is 70%.
With limited lung metastases and treatment with
Figure 15.9 chemotherapy plus surgery or radiotherapy
Management of Ewing’s sarcoma Euro Ewing 99 5-year survival of 20–40% can be expected. With
Protocol.
more extensive disease 5-year survival falls to less
than 25%.
Recurrent Ewing’s sarcoma
Radiotherapy Recurrence usually occurs within the first 2–5
Unlike osteosarcomas, Ewing’s tumours are radio years and the prognosis for these patients is poor.
sensitive and, radiotherapy used to be the prin Relapse after 5–10 years is associated with a
ciple method of achieving disease control. Studies survival similar to that of advanced disease at
have shown radiotherapy without surgery has a presentation.
worse outcome than surgery for local control but After 10 years very few patients relapse but it
there may be some selection bias. However, does occur. Treatment depends on the nature of
radiotherapy still remains a primary method of relapse (local/systemic), nature of initial chemo
local control in unresectable tumours when doses therapy and time from the end of primary treat
of 45–55 Gy have been used (Box 15.5). ment. Isolated lung metastases after a long
Management of metastatic disease treatment interval may be suitable for surgical
resection and further chemotherapy. To date
Some patients with limited pulmonary metas
high-dose regimens remain experimental in this
tases at presentation may still be cured with
group. The type of chemotherapy depends on the
combination treatment.
initial regimen but high-dose ifosfamide with
Chemotherapy etoposide has shown some activity. Trials of new
The same combination chemotherapy agents are agents such as insulin growth factor 1 receptor
used in advanced disease as in localized disease. antagonists are underway.
252
CANCERS OF THE MUSCULOSKELETAL SYSTEM 15
Chondrosarcoma
The most common sites are pelvis (Figures 15.10
and 15.11), axial skeleton and proximal limbs.
As with other bone tumours axial tumours have
a worse prognosis than extremity tumours. It
presents with a mass which may be painful or
painless or with symptoms due to local disease.
Systemic symptoms are rare. Diagnosis may be
suggested from plain X-rays but MRI should be
undertaken for suspected sarcomas.
Chondrosarcomas frequently exhibit calci
fication. MRI can also assess soft tissue exten
sion. The lesion often has a cauliflower
appearance. 5-year survival is over 90% for a
localized grade 1 (well-differentiated) tumour Figure 15.10
but reduces to 25% for a grade 3 (poorly dif X-ray of pelvis showing destructive lesion around right
ferentiated) tumour. pubic ramus (arrow).
253
II SITE-SPECIFIC CANCER MANAGEMENT
Figure 15.11
T1 weighted MRI of same patient showing extensive
chondrosarcoma (arrows) with some calcification and
soft tissue extension either side of the pubic ramus.
Pathology size >5 cm, deep to the deep fascia, rapid growth
There are over 80 subtypes of soft tissue sarcoma. and pain. Systemic symptoms (fever, weight
The most common are leiomyosarcoma, lipo loss, anorexia, breathlessness) may be present in
sarcoma, synovial sarcoma, rhabdomyosarcoma, advanced disease, particularly if associated with
fibrosarcomas (several subtypes), and malignant extensive chest disease.
peripheral nerve sheath tumour. Many soft tissue
Diagnosis
sarcomas are characterized and classified accord
ing to specific translocations and gene rearrange Correct diagnosis requires an adequate biopsy
ments (Box 15.6). which, like bone tumours, should not compro
mise later surgery. Samples should be sent to
Presenting features microbiology if infection is suspected. Pathology
Presentation depends on the stage and site of should be reviewed by an experienced sarcoma
disease as well as the histological subtype of pathologist. Due to the difficulty of diagnosis
sarcoma. The majority of soft tissue masses will even with immunohistochemistry, specialist tech
be benign but risk factors for malignancy include niques such as cytogenetics and polymerase chain
reaction (PCR) may also be necessary to identify
Box 15.6: Examples of soft tissue sarcomas translocation specific sarcomas.
which have characteristic chromosomal
translocations Staging
Translocations associated with sarcomas Staging should include evaluation of the primary
Type of fusion tumour and a CT of the chest to exclude lung
Translocation Genes gene
metastases. In certain rare subgroups (rhabdo
Ewing’s family of tumours
myosarcoma, synovial sarcoma, clear cell and
t(11;22)(q24;q12) EWSR1-FLI1 Transcription
factor epithelioid sarcomas) nodal disease may occur so
t(21;22)(q22;q12) EWSR1-ERG Transcription regional CT scanning may be appropriate. The
factor role of bone scintigraphy is less established in soft
t(7;22)(p22;q12) EWSR1-ETV1 Transcription
factor tissue sarcomas than bone sarcomas. PET has not
t(17;22)(q21;q12) EWSR1-ETV4 Transcription been fully evaluated (except for GIST) in soft
factor tissue sarcomas. Several staging systems exist,
t(2;22)(q33;q12) EWSR1-FEV Transcription
factor one of the most frequently used is AJCC (Table
Clear-cell sarcoma 15.2) which does not include histology and
t(12;22)(q13;q12) EWS-ATF1 Transcription site, and should not be used for non-extremity
factor sarcomas.
Desmoplastic small round-cell tumour
t(11;22)(p13;q12) EWSR1-WT1 Transcription General management principles of soft
factor
Myxoid chondrosarcoma tissue sarcoma
t(9;22) EWSR1-NR4A3 Transcription The overall management is shown in Figure
(q22-31;q11-12) factor
15.13.
Myxoid liposarcoma
t(12;16)(q13;p11) FUS-CHOP Transcription Localized disease
factor Surgery
t(12;22)(q13;q12) EWSR1-CHOP Transcription
factor Resection has to extend beyond the tumour to a
Alveolar rhabdomyosarcoma wide margin as an involved resection margin is
t(2;13)(q35;q14) PAX3-FKHR Transcription the most important factor which predicts risk of
factor recurrence. Margins have to be defined in terms
t(1;13)(p36;q14) PAX7-FKHR Transcription
factor of compartments and fascial or skin borders as
Synovial sarcoma well as distance. Surgery should be discussed
t(X;18)(p11;q11) SYT-SSX Transcription within a sarcoma multidisciplinary team to ensure
factor morbidity is minimized with the use of radio
therapy if required.
255
II SITE-SPECIFIC CANCER MANAGEMENT
Table 15.2: AJCC staging system for soft tissue sarcomas. This uses four criteria of tumour size,
nodal status, grade, and metastasis (TNGM)
Grade and TNM definitions Grade and TNM definitions
Tumour grade (G) AJCC Stage Groupings
GX Grade cannot be assessed Stage I
G1 Well differentiated Stage I tumour is defined as low-grade, superficial,
and deep
G2 Moderately differentiated
G1, T1a, N0, M0
G3 Poorly differentiated
G1, T1b, N0, M0
G4 Poorly differentiated or undifferentiated
G1, T2a, N0, M0
Primary tumour (T)
G1, T2b, N0, M0
TX Primary tumour cannot be assessed
G2, T1a, N0, M0
T0 No evidence of primary tumour
G2, T1b, N0, M0
T1 Tumour 5 cm or less in greatest
dimension: G2, T2a, N0, M0
• T1a: Superficial tumour G2, T2b, N0, M0
• T1b: Deep tumour
Stage II
T2 Tumour 5 cm or larger in greatest
Stage II tumour is defined as high-grade,
dimension:
superficial, and deep
• T2a: Superficial tumour
G3, T1a, N0, M0
• T2b: Deep tumour
G3, T1b, N0, M0
[Note: Superficial tumour is located exclusively
above the superficial fascia without invasion of the G3, T2a, N0, M0
fascia; deep tumour is located either exclusively
G4, T1a, N0, M0
beneath the superficial fascia, or superficial to the
fascia with invasion of or through the fascia, or G4, T1b, N0, M0
both superficial yet beneath the fascia.
G4, T2a, N0, M0
Retroperitoneal, mediastinal, and pelvic sarcomas
are classified as deep tumours.] Stage III
Regional lymph nodes (N) Stage III tumour is defined as high-grade, large,
and deep.
NX Regional lymph nodes cannot be assessed
G3, T2b, N0, M0
N0 No regional lymph node metastasis
G4, T2b, N0, M0
N1 Regional lymph node metastasis [Note:
Presence of positive nodes (N1) is Stage IV
considered stage IV.]
Stage IV is defined as any metastasis to lymph
Distant metastasis (M) nodes or distant sites.
MX Distant metastasis cannot be assessed Any G, any T, N1, M0
M0 No distant metastasis Any G, any T, N0, M1
M1 Distant metastasis
256
CANCERS OF THE MUSCULOSKELETAL SYSTEM 15
Figure 15.13
Biopsy proven
Management of limb soft tissue
sarcomas (LG-low grade; HG-high
grade).
Staging
MRI of primary if not done
CT chest
Other imaging according to specific
subtype e.g. bone scan, regional CT/
Assess for surgery
Metastatic Localized
Painful metastases
Radiotherapy tumour bed but must not cross the fascial com
Surgery is the primary curative modality for soft partment to avoid lymphoedema and delayed
tissue sarcomas; radiotherapy has a significant fibrosis which significantly affects mobility.
role in the prevention and treatment of local Chest wall sarcomas may be close to the heart
recurrence although there is no evidence that or spinal cord making planning complex and
radiotherapy improves overall survival. Many potentially limiting doses delivered. They may
soft tissue sarcomas are radiosensitive. A dose of follow the pleural contour of the chest wall
>60 Gy is needed and the radiotherapy details making it difficult to avoid significant lung
are shown in Box 15.5. volumes within the treatment field.
For some patients who are unfit for surgery Preoperative and postoperative radiotherapy
or in whom resection is technically not feasible, Radiotherapy is usually planned postoperatively
radiotherapy may be considered as a primary in two phases to take into account possible
treatment modality. However, unless the tumour tumour spillage at the edge of the surgical pro
is very small and superficial, local control will cedure then to focus on the central tumour bed.
not be as good as with surgery. Planning must take into account preoperative
Limb sarcoma radiotherapy presents several scans to estimate tumour location, but should
challenges including positioning and immobiliz also encompass the surgical edges. Preoperative
ing the limb to ensure consistent placement and radiotherapy allows a single phase approach.
accurate dose to the tumour bed with a margin. An ongoing trial, VORTEX, is examining the
An adequate dose must be delivered to the possibility of reducing the volume of the post
257
II SITE-SPECIFIC CANCER MANAGEMENT
operative treatment comparing a single phase surgeons. Radiotherapy is recommended for the
treatment with a standard 2-phase approach. majority of cases except low-grade tumours
Radiotherapy has traditionally been given which have been excised with a wide margin.
postoperatively but the potential benefits of Radiotherapy is indicated for low grade tumours
giving preoperative radiotherapy include a with involved resection margin when further
smaller treatment volume, tumour visible on surgery is not contemplated. The role of adjuvant
planning scan, reduction of tumour prior to chemotherapy for most subgroups remains con
surgery allowing a less morbid and complete troversial but could be considered within the
operation and reduction in significant late toxic context of a clinical trial (Box 15.8).
ity such as limb oedema, joint restriction and
Advanced and metastatic soft tissue sarcomas
fibrosis. Concerns about using preoperative radi
otherapy are effect it may have on interpretation Many sarcomas present with locally advanced or
of histology and its associations with wound metastatic disease. In the majority of cases this
complications postoperatively. presentation carries a very poor prognosis. Prog
nostic factors are listed in Box 15.9. The median
Adjuvant chemotherapy survival with metastatic disease is 12–14 months.
The use of adjuvant chemotherapy in soft tissue In other cases, presentation with limited
sarcomas remains controversial and is com metastatic disease occurs several years after their
pounded by different histological subtypes of initial treatment for localized sarcoma and the
sarcoma with varied chemo-sensitivities being prognosis may be slightly better in this situation,
included as a group in the majority of studies. if the disease is operable. In all these situations
There are some sarcomas, which tend to pre treatment options depend on performance
dominate in the paediatric setting, in which adju status, co-morbidities and histological subtype of
vant treatment has been shown to be useful, sarcoma.
e.g. rhabdomyosarcomas and the extraskeletal
Ewing’s sarcoma. However, for the majority Chemotherapy
there is no consensus concerning the use of adju Systemic treatment with chemotherapy can be
vant chemotherapy (Box 15.7). A meta-analysis useful but its role is more complex than in other
showed a statistically significant improvement in malignancies. There is little evidence that chemo
local and distant recurrence if adjuvant chemo therapy prolongs survival but it can provide sig
therapy was given. There was a trend towards an nificant benefits in symptom control. Response
improved overall survival of 4% at 10 years, but rates as determined by RECIST criteria often do
this was not statistically significant. A multi-cen not reflect symptomatic benefit or disease control,
tre international EORTC study randomized 351 especially in response to certain agents such as
patients to standard treatment versus ifosfamide trabectedin. Different tumour types show differ
with doxorubicin. A preliminary analysis has ent responses to chemotherapy in general and to
shown no statistically significant difference specific drugs (Box 15.10).
between the two groups in either local recurrence Doxorubicin is a standard first agent with
rates or overall survival. response rates of 20–25%. Doses below 60 mg/m2
Treatment recommendations
Surgical resection with wide margins remains the
mainstay of treatment for soft tissue sarcoma. Box 15.8: Sensitivity to chemotherapy of soft
This should be performed by specialist sarcoma tissue sarcomas
The most chemosensitive tumours are synovial
sarcoma, myxoid liposarcoma and childhood
rhabdomyosarcoma. Extraskeletal Ewing’s tumours,
Box 15.7: Adjuvant chemotherapy
discussed under bone tumours, are also chemo-
The use of adjuvant chemotherapy is not routinely sensitive. The most chemoresistant tumours are
recommended outside a clinical trial, but the studies dedifferentiated liposarcoma, alveolar soft part
should be discussed with patients along with their sarcoma, GIST, low-grade liposarcoma and clear cell
particular situation. sarcoma.
258
CANCERS OF THE MUSCULOSKELETAL SYSTEM 15
Box 15.9: Prognostic factors in soft tissue Box 15.11: When to use the combination of
sarcomas ifosfamide and doxorubicin
Different prognostic factors predict for local and There are a few special circumstances in which
systemic recurrence: combined chemotherapy may be used first line:
• Size and grade best predict distant recurrence and • Life-threatening progressive disease
overall survival. • Preoperatively to downstage and aim for
• Other factors which predict survival include age, resection
anatomical site and histological subtype. • Unassessable advanced disease (e.g. diffuse
• Retroperitoneal and visceral tumours carry a worse peritoneal metastasis)
prognosis than extremity tumours as, like pelvic
Ewing’s, they can grow to a very large size before
they are detected.
• Specific tumour subtypes such as MPNST and Box 15.12: Criteria for resection of lung or
leiomyosarcoma have a poorer overall survival. intra-abdominal metastases in metastatic soft
• Increased proliferative activity as assessed by tissue sarcoma
immunocytochemical stains has shown a • No other sites of disease, including primary site
correlation with poorer survival in at least three
studies. • A long disease-free interval of at least a year
• In synovial sarcomas, several studies have shown a • The disease should be slowly progressing or stable
poorer prognosis with the SYT-SSX1 combination • Complete resection must appear possible
rather than SYT-SSX, but this has not been • Low numbers of metastases although some centres
confirmed in other studies. have removed multiple metastases
• Involved resection margins and age >50 are worst
for local recurrence.
• Specific histological tumour types with a higher
rate of local recurrence include MPNST and with no improvement in PFS or overall survival
fibrosarcomas. and with increased toxicity. Combination chemo
• Nomograms have been developed, and validated, therapy does have a role in the childhood and
which help to predict survival based on these
features. The best known is the MSK nomogram adolescent tumours such as rhabdomyosarcoma
which can be found at http://www.mskcc.org/ and extraskeletal Ewing’s sarcoma (Box 15.11).
mskcc/html/443.cfm
Surgery
Despite the poor median overall survival with
metastatic disease a small group of patients have
Box 15.10: Specific chemotherapy and histology a prolonged survival most of whom have disease
Recent studies have shown that some chemotherapy which is amenable to surgery, e.g. lung metas
agents are more effective in some tumours: tases or isolated intra-abdominal recurrence. No
• Taxanes and liposomal doxorubicin have been randomized trial has evaluated the benefit of sur
shown to be effective in angiosarcomas.
gical resection of lung metastases but the 5-year
• Gemcitabine and docetaxel initially showed
activity in uterine leiomyosarcomas but also have survival of patients selected using strict criteria
a response rate up to 30% in other (Box 15.12) is up to 40%.
leiomyosarcomas. Resection of liver metastases, other than for
• Trabectedin has been shown to be effective in the GIST, is performed rarely and there is little
treatment of liposarcomas, especially myxoid
liposarcomas. It also has over a 30% response rate evidence that it is of benefit either in terms of
in previously treated leiomyosarcomas and some survival or symptom control.
activity in other sarcomas.
Radiotherapy
The majority of soft tissue sarcomas are radio
sensitive and radiotherapy can be an effective
have been shown to be ineffective and above tool in symptom control. Doses used are in the
75 mg/m2 to have increased toxicity without clini range of 35–54 Gy.
cal benefit. Ifosfamide is often used as a second
line agent and has a response rate of 25% overall. Isolated limb perfusion
In general combination chemotherapy has been Some patients present with a massive inoperable
shown to improve response rates up to 46% but limb sarcoma, often accompanied by significant
259
II SITE-SPECIFIC CANCER MANAGEMENT
Retroperitoneal sarcoma
Retroperitoneal sarcomas represent 13% soft
tissue sarcomas. The most common histologies
at this site are liposarcoma (Figure 15.14) and
leiomyosarcoma. Many are found incidentally.
When they occur symptoms are due to a large
mass (abdominal swelling, leg swelling distal to
the mass, pain).
If operable a biopsy may not be performed and
the tumour resected en masse to prevent tumour
seeding the biopsy tract. If not operable a biopsy
must be obtained to ensure correct management
of an unsuspected alternative diagnosis. Suffi
cient material must be obtained to perform
immunohistochemistry to exclude lymphoma,
carcinoma and germ cell tumours. Figure 15.15
Adjuvant chemotherapy is not standard treat Large gastric GIST (shown by arrows) arising from the
ment in these tumours and in one retrospective greater curvature of the stomach.
261
II SITE-SPECIFIC CANCER MANAGEMENT
263
Central nervous
system tumours
TV Ajithkumar
16
Introduction 2. Prior radiotherapy increases the risk of a second
intracranial tumour, particularly meningiomas
Primary central nervous system (CNS) tumours and gliomas with a typical latent period of 10
can arise from any structures in the cranial vault. years. Immunosuppression increases the risk of
Around 4000 new patients with malignant brain primary CNS lymphoma.
tumours are diagnosed per year in the UK and 3. Brain tumours are not associated with
22,000 new patients in USA. There is a bimodal smoking, alcohol intake or mobile phone use.
distribution with small peak in children (p. 323)
and a steady increase starting at the age of
20 years. Males are more commonly affected, Pathology
particularly with malignant tumours, whereas Box 16.1 shows WHO classification of brain
women have a higher rate of non-malignant tumours. WHO grading system (Box 16.2) is
tumours, particularly meningiomas. important in deciding management and progno-
sis. Molecular features can be incorporated in
Aetiology this grading system to yield important prognostic
information. Clinico-pathological features of
The majority of brain tumours are sporadic with individual tumours are discussed later.
an unknown cause. A small proportion of
tumours are associated with genetic syndromes
and other factors. Presentation
1. Genetic syndromes – fewer than 5% patients Brain tumour can present with non-specific as
with glioma have a family history of brain well as specific features. Non-specific features
tumours: are headache (50%) and vomiting due to raised
• Neurofibromatosis type I – neurofibro- intracranial pressure. Specific features are due
mas, optic nerve glioma and pilocytic to location of the tumour. Lateralizing signs
astrocytoma. (50%) include hemiparesis, aphasia and visual
• Neurofibromatosis type II – meningioma, field defects. Seizure, generalized, partial or
gliomas and bilateral acoustic neuroma. focal, can be the presenting symptom in 50% of
• Tuberous sclerosis – subependymal giant high-grade and 25% of low-grade gliomas.
cell astrocytoma, hamartomas. Stroke-like presentations occur mainly due to
• Turcot syndrome – glioblastoma and bleeding associated with the tumour. It can occur
medulloblastoma. in 5–8% of high-grade and 7–14% of low-grade
• Li–Fraumeni syndrome – astrocytoma and tumours. Altered mental status can be a present-
PNET. ing feature of primary brain tumour, but is
• Basal naevus syndrome – medulloblast- more commonly a presentation of multiple brain
oma. metastases.
264 © 2011, Elsevier Ltd
DOI: 10.1016/B978-0-7234-3458-0.00021-X
CENTRAL NERVOUS SYSTEM TUMOURS 16
Box 16.1: WHO classification of brain tumours Box 16.2: WHO grading of primary CNS tumours
Tumours of neuroepithelial tissue Grade 1 – tumours with low proliferative potential
1. Astrocytic tumours and tumour growth by expansion. Generally
curable by surgery alone.
• Pilocytic astrocytoma (grade 1)
Grade 2 – tumours with low proliferative potential
• Diffuse astrocytoma (grade 2) but exhibit infiltration. Surgery alone is rarely
• Anaplastic astrocytoma (grade 3) curative and these tumours can progress to grade
• Glioblastoma (grade 4) 3–4 tumours.
2. Oligodendroglial tumours Grade 3 – tumours with cytological evidence of
malignancy such as nuclear atypia, endothelial
• Oligodendroglioma (grade 2) proliferation and mitotic activity. These are rarely
• Anaplastic oligodendroglioma (grade 3) curable.
3. Ependymal tumours Grade 4 – highly malignant tumours often with
• Ependymoma (grade 2) necrosis. These are aggressive.
• Anaplastic ependymoma (grade 2)
4. Mixed glioma
• Oligoastrocytoma (grade 2)
• Anaplastic oligoastrocytoma (grade 3)
Choroid plexus tumours oedema, hydrocephalus, haemorrhage and calci-
• Papilloma and carcinoma fication depending on the histological variant of
Embryonal tumours brain tumour. Table 16.1 shows radiological
• Medulloblastoma and its variants (grade 4) appearance of common tumours.
• Supratentorial primitive neuroectodermal
tumour (PNET) (grade 4) MRI scan
Pineal tumours MRI scan with gadolinium and FLAIR (fluid-
Neuronal tumours attenuated inversion recovery) sequence is the
• Ganglioglioma standard investigation for brain tumours. T1W
• Anaplastic ganglioglioma imaging demonstrates anatomy and areas of con-
• Neurocytoma trast enhancement and T2W and FLAIR images
Tumours of cranial/spinal nerves are useful in demonstrating oedema. Appearance
Tumours of the meninges of tumour on T1W image is similar to that on
• Meningioma (grade 1–3) CT, but better delineated on MRI. Tumour and
Tumours of uncertain histogenesis oedema demonstrate increased signal on T2 and
Haematopoietic neoplasms the area of increased T2 signal on MRI usually
Germ cell tumours includes the hypodense area on CT (Figure 16.1).
Tumours of sellar region Various MR imaging techniques are being
• Pituitary adenoma and carcinoma evolved to improve the diagnostic ability in the
• Craniopharyngioma imaging of brain tumours.
Metastatic tumours
Magnetic resonance spectroscopy (MRS)
MRS is clinically useful in distinguishing high-
grade glioma and metastases from abscess,
assessing tumour recurrence and radiation necro-
Investigations sis, and identification of atypical meningioma as
a true meningioma.
Imaging
Diffusion tensor imaging (DTI)
CT scan In radiotherapy planning for high-grade gliomas,
Contrast enhanced CT scan is usually the initial conventional methods of imaging cannot distin-
investigation in patients suspected to have a guish oedema from peritumoural white matter
brain lesion. CT scan is not an ideal investigation infiltration, resulting in large target volumes. DTI
for low-grade tumours or tumours in the poste- shows white matter abnormalities resulting from
rior fossa. CT scan may also show features of tumour infiltrating, and thereby reducing the
265
II SITE-SPECIFIC CANCER MANAGEMENT
target volume, which would allow significant moma (Figure 16.2). CSF evaluation includes
dose escalation to the tumour with acceptable CSF biochemistry (typically elevated protein
damage to the normal tissue. >40 mg/dL and reduced glucose <50 mg/mL),
cytology and in cases of suspected germ cell
Evaluation of the craniospinal axis tumours, the estimation of tumour markers (AFP
MRI of the craniospinal axis and CSF evaluation and beta hCG) in CSF and serum. Evaluation of
is needed for tumours with a high risk of CSF the craniospinal axis is best done prior to surgery
dissemination such as medulloblastoma, germ or >3 weeks after surgery to avoid false positive
cell tumours, CNS lymphoma, PNET and ependy results.
266
CENTRAL NERVOUS SYSTEM TUMOURS 16
A B C
Figure 16.1
MRI scan of diffuse astrocytoma. Scan shows hypointense lesion on left parieto-occipital region (A) without contrast
enhancement (B) on T1W image. T2W image (C) shows a hyperintense lesion. Since low-grade tumours are not
associated with oedema, this hyperintense lesion represents just the tumour.
Prognostic factors
The outcome of brain tumours depends on the
following factors:
1. Pathology – outcome depends on pathologic
type and grade. High-grade tumours have
poor prognosis. Recently molecular factors
are shown to be important in certain tumour
types.
2. Age – increased age is associated with poorer
outcome.
3. Performance status – is an important factor
in outcome and guiding treatment.
4. Extent of surgical resection – based on
observational studies, patients with less
residual disease after surgery have a better
Figure 16.2 prognosis.
Multiple contrast-enhancing tumour nodules in
ependymoma suggesting leptomeningeal disease.
Principles of management
Tissue diagnosis General medical management
Histologic confirmation is necessary in all patients Steroids
prior to definite treatment. Biopsy is obtained Steroids may improve symptoms by reducing
by either stereotactic guided techniques or open intracranial pressure. The most commonly used
biopsy. In intrinsic brainstem tumours, biopsy agent is dexamethasone which is started at a dose
267
II SITE-SPECIFIC CANCER MANAGEMENT
of 2–16 mg daily (low doses given once daily and guide resection based on real time shift of
higher doses as 2–4 equally divided doses) and structures during operation.
titrated against the patient’s symptoms. The 3. Cortical mapping awake craniotomy – is
optimal dose is just above that at which sympto- useful in resection of tumours inside or adja-
matic deterioration occurs. If there is no symp- cent to functional brain areas. Intraoperative
tomatic improvement, steroids should be stopped. cortical or subcortical stimulation helps to
map functional areas. Initial craniotomy and
Anti-epileptics preliminary stimulation are done with the
All patients who have had a seizure should patient asleep. After arousal, under sedative/
be treated with an anti-epileptic drug (AED); hypnotic anaesthesia, the patient responds to
prophylactic use of anti-epileptics is not recom- motor or language commands but with sub-
mended. Many anti-epileptics are enzyme- sequent amnesia.
inducers, and hence should be carefully monitored 4. Fluorescent guided surgery – an evolving tech-
when they need to be used concurrently with nique for malignant gliomas.
drugs metabolized by cytochrome p450 enzyme
system. Radiotherapy in brain tumours
Analgesics Radiotherapy has a major role in the manage-
ment of CNS tumours. Radiotherapy is proven
Patients requiring pain control are managed
to improve local control and/or survival in grade
according to the WHO analgesic ladder (p. 66).
3–4 tumours whereas its role in grade 2 tumours
Surgical management is doubtful and has limited role for grade 1
Surgery in brain tumours helps in pathological tumours. Radiotherapy is delivered by the fol-
diagnosis, symptom control and definite lowing methods:
treatment. 1. External beam radiotherapy is commonest
Biopsy can be either stereotactic guided or mode of delivery of radiotherapy to the brain.
an open biopsy. Surgery is useful in relieving Immobilization and proper localization is
pressure symptoms as well as decreasing the important in the delivery of EBRT. Target
frequency and intensity of seizures. Surgical volume depends on the type of tumour and
resection is constantly evolving in brain tumours. intention of treatment.
It is common practice to confirm the diagnosis 2. Stereotactic technique – this technique involves
with an intraoperative frozen section or smear improved localization of tumour and preci-
prior to attempting a full resection. Complete or sion. Treatment is delivered either as multiple
maximal resection with minimal injury to neigh- fractions using linear accelerator (called
bouring critical structures is the aim of surgery. stereotactic radiotherapy) or as single fraction
The importance of surgical resection in specific (called radiosurgery) using gamma knife, a
tumours is discussed later. The various resection special machine for cranial radiosurgery.
techniques are: 3. Charged particle radiotherapy – mainly
1. Image guided stereotactic resection – is useful protons, which deposit the energy only at a
in technically challenging locations such as depth and hence useful in skull base tumours.
the peri-thalamic region, to minimize critical A summary of radiotherapy details for
structure injury. common tumours are given in Table 16.2.
2. Neuronavigation guided resection – distor-
tion of brain anatomy by tumour and oedema Radiation tolerance and reactions
makes surgical resection according to conven- Radiation to the brain can cause structural
tional landmarks difficult. Neuronavigation damage to critical structures if attention is not
systems allow registration of stored imaging paid not to exceed the tolerance dose of indi-
data which can be incorporated with real time vidual structures.
intra-operative imaging using intra-operative Acute radiation reactions may manifest up to
ultrasound or MRI. This helps surgeons to 6 weeks following completion of irradiation.
268
Table 16.2: Radiotherapy for common brain tumours
Tumour Intent of treatment GTV* CTV+ (margin added to GTV) Dose
Low-grade glioma Radical Abnormality on T2W image on fused image 1.5 cm 54 Gy in 30#
Grade III glioma Radical Contrast enhancing abnormality on co-registered Phase I – 2.5 cm 45 Gy in 25#
image Phase II – 1.5 cm 9 Gy in 5#
Grade IV glioma Radical Contrast enhancing abnormality on co-registered Phase I – 2.5 cm 50 Gy in 25#
image Phase II – 1.5 cm 10 Gy in 5#
Palliative Contrast enhancing tumour 2.5 cm 30 Gy/6# or
35 Gy/10#
Brainstem glioma Radical Abnormality on T2W/FLAIR on co-registered image 1–1.5 cm 54 Gy/33#
Medulloblastoma Adjuvant CT planning for craniospinal RT Phase I – whole craniospinal axis 35 Gy/21#
Abnormality on fused image for phase II Phase II – posterior fossa or visible 20 Gy/12#
tumour + 1.5 cm
Phase II – spinal metastasis boost 15 Gy/9#
Ependymoma Adjuvant/radical Contrast enhancing lesion or surgical cavity in case 1.5–2.5 cm 55 Gy/33# or
of complete resection on fused image 54 Gy/30#
Germinoma Radical Phase 1 – CT planning Phase I – whole craniospinal axis 25 Gy/15#
Phase 2 – primary and other metastatic sites on Phase 2 – plan before start of 15 Gy/9#
fused image treatment. Contrast enhancing
tumour + 1–2 cm
Non-germinoma After chemotherapy CT planning Phase 1 – craniospinal RT 30 Gy in 20#
– meningeal disease
Abnormality on co-registered image Phase 2 primary tumour + 2 cm 24 Gy/12#
Residual disease Contrast enhancing mass on co-registered image Primary + 2 cm 54 Gy/33#
Acoustic neuroma Progressive disease Residual tumour on fused image No margin needed 50 Gy/30#
Craniopharyngioma Postoperative/ Residual tumour on fused image Preoperative GTV + margin for 50 Gy/30#
progressive doubtful margin
Pituitary adenoma Postoperative or Visible tumour on co-registered image No margin 45 Gy/25#
progressive
Meningioma Radical/postoperative Contrast enhancing tumour on co-registered 0–0.5 cm 50 Gy/30# or
image. The practice of including dural tail in 55 Gy/33#
GTV is variable; many studies fail to show any
meningioma in dural tail
*GTV is outlined on MRI co-registered with planning CT scan. T2W MRI is used for co-registration in low grade glioma, whereas contrast enhanced T1W is used for high-grade glioma,
meningioma, craniopharyngioma and pituitary adenoma.
+CTV needs to be edited to confine to skull or skull base. PTV depends on immobilization device: for Perspex shell add 5 mm to CTV and for relocatable stereotactic frames add 2–3 mm to
CENTRAL NERVOUS SYSTEM TUMOURS
CTV.
Normal tissue tolerance doses (at ≤2 Gy per fraction): brain 60 Gy; brainstem 54 Gy; optic nerve and chiasm 50–54 Gy; pituitary gland <40 Gy; middle and inner ear <40 Gy; lacrimal gland
<20 Gy; lens <10–15 Gy as fractions; permanent alopecia 43 Gy.
16
269
II SITE-SPECIFIC CANCER MANAGEMENT
These reactions are generally those of raised Box 16.3: Chemotherapy regimes in
intracranial pressure, fatigue and worsening of brain tumour
neurology. Vomiting is particularly common in PCV (glioma)
patients receiving radiotherapy to the brainstem • Procarbazine 100 mg/m2 (max 200 mg) oral Day
region. 1–10
Early delayed (intermediate) side effects usually • CCNU 100 mg/m2 (max 200 mg) oral Day 1
appear from 6 weeks to 6 months following • Vincristine 1.5 mg/m2 (max 2 mg) IV Day 1
radiotherapy and are due to changes in capillary Cycles repeated every 6 weeks for 6 cycles
permeability and transient demyelination. Symp- Temozolomide (glioma)
toms include headache, somnolence syndrome, • Concomitant – 75 mg/m2 D1–D42 oral, 1 hour prior
to radiotherapy & morning dose on weekends
lethargy and frequently recurrence of the original
• Adjuvant and palliative – 150 mg/m2 D1–5 oral for
presenting features. Recovery is spontaneous but first cycle, then 200 mg/m2 D1–D5 total 6 cycles
can be accelerated by steroids. (adjuvant starting 4 weeks after radiotherapy)
Delayed radiation reaction develops 6 months Packer regime (medulloblastoma and PNET)
after radiotherapy. This is thought to be due to • Concurrent with radiotherapy – vincristine 1.5 mg/
white matter injury secondary to vascular injury, m2 (max 2 mg) weekly with radiotherapy, then 6
weeks of rest followed by:
demyelination and necrosis. The most serious
• A combination of:
form of this is radiation necrosis which peaks at
• CCNU 75 mg/m2 every 6 weeks and cisplatin
3 years. Conventional imaging fails to distinguish 68 mg/m2 every 6 weeks and vincristine 1.5 mg/
this from recurrent tumour and special imaging m2 (max 2 mg) for 3 consecutive weeks. Cycle
is therefore needed (p. 265). Treatment is with repeated every 6 weeks for total of 8 cycles
steroids and debulking. Other late effects include,
depending on the area irradiated, memory prob-
lems, pituitary failure, hearing loss, visual changes
and second malignancies.
antagonists (e.g. imatinib) and farnesyltrans-
Chemotherapy in brain tumours ferase inhibitors (e.g. tipifarnib).
Chemotherapy has an established role in paedi-
atric brain tumours mainly as a substitute for Follow-up
radiotherapy. The role of chemotherapy in adult
Follow-up consists of clinical evaluation with
brain tumours is increasing. The active agents are
attention to neurological examination and assess-
nitrosoureas (carmustine and lomustine), procar-
ment of seizure control. MRI is the image of
bazine, temozolomide, platinum and vincristine.
choice whenever imaging follow-up is indicated.
The role of chemotherapy in specific brain
Visual field and hormonal function assessments
tumours is discussed later in this chapter and the
are important in sellar tumours.
commonly used chemotherapy regimens are
given in Box 16.3.
Clinico-pathological features
New agents in brain tumours and management of
A number of biological agents have been tried specific tumours
recently in the management of brain tumours,
particularly in gliomas. Since gliomas exhibit Glioma
marked neovascularization, anti-angiogenesis Astrocytomas diffusely infiltrate surrounding
inhibitors are an attractive option. Bevacizumab brain. Pilocytic astrocytoma (grade 1) occurs in
has been studied alone and in combination with children and young adults and are located in the
chemotherapy in recurrent GBM. optic tract, hypothalamus, basal ganglia and pos-
Since EGFR are frequently amplified and over- terior fossa. These tumours grow slowly and sta-
expressed in GBM, agents such as cetuximab and bilize spontaneously. Diffuse astrocytoma (grade
tarceva seem to be attractive agents. Other target 2) is a low-grade tumour. Anaplastic astrocy-
molecules under investigation include PDGF toma (grade 3) and glioblastoma multiforme
270
CENTRAL NERVOUS SYSTEM TUMOURS 16
A B
Figure 16.3
MRI scan of oligodendroglioma in right temporal area. MRI scan of oligodendroglioma will exhibit the same features
of astrocytoma (A); however oligodendroglioma can have subtle contrast enhancement (arrows on B) and calcification.
(grade 4-GBM) are high grade tumours. Ana tumours with progressive neurological deficit
plastic astrocytoma is distinguished from low- are treated with radiotherapy and carboplatin-
grade tumours by greater cellular differentiation based chemotherapy.
and hyperchromasia, more frequent mitoses and
prominent small vessels lined by epithelioid Treatment of low grade glioma
endothelial cells. Glioblastoma is distinguished (WHO grade 2)
by increased cellularity, pleomorphism, giant Diffuse astrocytoma, oligodendroglioma (Figure
cells, abnormal mitotic figures and endothelial 16.3) and mixed oligoastrocytoma are grouped
cell proliferation. Necrosis surrounded by nuclei together as low-grade glioma. These tumours
aligned in pseudopalisading pattern is character- mainly affect the young adults (mean age of 35
istic. Astrocytomas stain for glial fibrillary acidic years for astrocytoma and 45 years for oligoden-
protein (GAFP). droglioma). Patients generally present with sei-
Oligodendroglial tumours have typical histo- zures and in the majority adequate seizure control
logical appearance of evenly distributed cells with with serial MRI monitoring will be sufficient.
uniform and rounded nuclei with perinuclear Many of these patients have slow growing
halo. Mixed tumours are composed usually of tumours. There are two main patterns of
an oligodendroglial component and an astro- progression – secondary gliomatosis cerebri
cytic component. Their overall prognosis and and malignant transformation. Secondary
treatment is dictated by the most aggressive gliomatosis cerebri is characterized by diffuse
component. extension of the tumour sometimes to the oppo-
site hemisphere and it often remains low-grade.
Treatment of pilocytic astrocytoma
Malignant transformation involves progression
Complete surgical resection alone is adequate to a high-grade tumour with characteristics of
which results in a 20-year survival of 80%. In anaplastic astrocytoma, anaplastic oligodendro-
patients for whom a complete resection is not glioma or glioblastoma. Imaging shows new
possible, maximal safe resection should be areas of irregular contrast enhancement in a pre-
attempted followed by observation. In the viously non-contrast enhancing tumour.
majority of these patients the tumour does not
progress. A second surgery may be attempted Surgery
during progression, if feasible, followed by non- The role of surgery in grade 2 tumours is unclear.
surgical management. Patients with unresectable When safe complete or near complete resection
271
II SITE-SPECIFIC CANCER MANAGEMENT
A B
Figure 16.4
GBM recurrence. GBM can spread along the white matter track and manifest with recurrence away from the original
lesion. This patient, who had left temporal lobectomy (B-long arrow) for GBM (A), presents with a non-homogenous
contrast enhancing recurrent lesion (B-arrow head), which indicates spread along the white matter track.
years and of enhancing lesions is approximately These are cured with excision. Choroid plexus
11 months. carcinomas (grade 3) are also common in chil-
dren and behave aggressively with extensive ven-
Ependymoma tricular and subarachnoid space spread. Choroid
Ependymomas (grade 2) are slow growing plexus carcinoma is treated with a combination
tumours arising from the ependymal or sub- of surgery and radiotherapy, with or without
ependymal cells surrounding the ventricles, spinal chemotherapy.
canal or filum terminale. These are typically low-
grade but exhibit a high risk of recurrence and Medulloblastoma and PNET
can spread through the CSF. They are rarely
Medulloblastoma (grade 4) commonly occurs in
infiltrative and seldom metastasize outside CNS.
children, but can also occur in adolescents and
Subependymoma, which occur in the cerebral
adults. It usually affects the cerebellum and can
ventricles and myxopapillary ependymoma,
spread locally, through CSF and outside the
which occurs in the filum terminale are grade 1
CNS. Histologically the tumour consists of small
variants of ependymoma. Histologically these
primitive round or spindle cells which might
tumours consist of uniform round cells with
express neuron specific enolase, synaptophysin,
widespread pseudorosette formation. Maximal
Leu-7 and protein gene product 9.5.
surgical resection followed by postoperative
PNET (grade 4) are aggressive supratentorial
radiotherapy is the treatment of choice in adults
tumours in children and young adults. Histology
(Table 16.2). Local radiotherapy is recommended
shows highly cellular diffuse sheets of cell with
if spinal MRI and CSF cytology are clear.
extensive necrosis. The prognosis is worse than
Craniospinal RT is recommended only in cases
medulloblastoma.
of neuraxis spread. There is no recommended
role for chemotherapy, except in recurrence
after radiotherapy. Postoperative radiotherapy Treatment of medulloblastoma and PNET
increases 5-year survival from 18% to 68%. In medulloblastoma, the extent of surgical resec-
tion correlates with survival and hence, maximal
Choroid plexus tumour surgical resection is attempted in localized
Choroid plexus papilloma (grade 1) usually disease. All patients are treated with postop
presents as an intraventricular mass in children. erative craniospinal radiotherapy followed by
274
CENTRAL NERVOUS SYSTEM TUMOURS 16
additional radiotherapy to the posterior cranial Germ cell tumours
fossa. Radiotherapy with concurrent vincristine Germ cell tumours occur in the midline struc-
followed by adjuvant chemotherapy with cispla- tures, pineal, sellar, hypothalamic and third
tin, CCNU and vincristine (Packer regime) is an ventricular regions. The majority of patients are
effective regime (Box 16.3); but it is very toxic diagnosed between 11–20 years. Histological
in adults (especially neurotoxicity) and hence appearance is similar to their gonadal counter-
sometimes radiotherapy alone may be used. part. Immunohistochemically germinomas may
stain for placental alkaline phosphatase (PLAP)
Acoustic neuroma and hCG and non-germinoma may stain for hCG
(vestibular schwannoma) and AFP.
Acoustic neuromas are tumours arising from the Presentation depends on the tumour type and
Schwann cell of the myelin sheath of the eight location. These tumours usually arise from the
cranial nerves. These can occur sporadically (4– pineal region and present with features of CSF
5th decade of life) or as part of NF2 (2nd–3rd obstruction and Parinaud’s syndrome (dorsal
decade of life). These are expansile slow growing midbrain compression leading to failure of up
tumours. Clinical presentation is usually with gaze, nystagmus and light-near dissociation of
progressive sensorineural hearing loss (up to pupil). Suprasellar tumours present with neuro
95%). Large cerebello-pontine angle tumours endocrine deficits.
can lead to trigeminal involvement and pressure On imaging germinomas appear as a homog-
effect on the cerebellum and brainstem can give enously enhancing mass and non-germinomas as
rise to ataxia and lower cranial nerve (IX–XII) a heterogeneous mass with cysts, calcification
involvement. and/or haemorrhage. Spinal imaging with MRI
Imaging shows a homogenously enhancing may show leptomeningeal disease in 10–15% of
lesion close to internal auditory canal. Follow-up cases.
with regular imaging may be adequate for some The role of surgery is mainly to obtain a
patients. When treatment is indicated, for pro- tissue biopsy. In tumour marker positive disease
gressive tumours or large tumours with pressure (AFP above 25 IU/L or serum or CSF hCG
symptoms, the treatment options are surgery and of >50 IU/L in Europe and >100 IU/L in
radiotherapy (either radiosurgery or fractionated USA), imaging alone is sufficient prior to non-
radiotherapy). Surgery achieves a complete exci- surgical treatment. Surgery may have a role in
sion of 97% at the expense of complete hearing symptomatic residual masses after non-surgical
loss and transient facial palsy. Radiosurgery is management.
appropriate for lesions of <3 cm size. Fraction- Germinoma is treated with craniospinal radio-
ated radiotherapy may preserve hearing in therapy, which results in a 5-year survival of
patients with good control rates; but long-term 90–95% and 10-year survival of 91%. Chemo-
follow-up is needed. therapy is being evaluated to avoid the need
for craniospinal radiotherapy and to reduce the
dose of radiotherapy in paediatric germinoma.
Pineal tumours
Non-germinoma is treated with platinum based
Pineal tumours are common in children. Pineocy- chemotherapy followed by radiotherapy. In non-
toma (grade 2) are slow growing tumours which germinoma, with radiotherapy alone the overall
are treated with surgery and radiotherapy. survival is only 10–30%, whereas combined
Pineoblastomas (grade 4) are aggressive tumours modality treatment results in a 5-year survival
treated with surgery, radiotherapy and chemo- of 45%.
therapy. Pineal parenchymal tumours of inter
mediate differentiation have an unpredictable
clinical behaviour. Treatment recommendation Meningeal tumours
varies from surgery alone to surgery followed by Meningiomas (Figure 16.5) account for 30% of
craniospinal radiotherapy. Management of pineal primary intracranial neoplasms and occur pre-
germ cell tumours is discussed below. dominantly in women. Grade 1 meningiomas
275
II SITE-SPECIFIC CANCER MANAGEMENT
A B
Figure 16.5
CT scan of meningioma. A, A homogenously contrast enhancing lesion arising from the dura. B, Bone window showing
increase in thickness of skull bone (hyperostosis) at the site of the tumour (arrows), a feature of meningioma.
(90% of meningiomas) are benign with a low apy is indicated (e.g. tumour near optic
risk of recurrence, atypical (grade 2) meningi- chiasma).
omas (5–7%) have a high risk of local recurrence 2. Grade of tumour – grade 2 and 3 tumours
after complete excision and grade 3 meningiomas have a 5-year recurrence rate of 40–100%
(3–5%) are aggressive. Papillary meningioma even after complete resection whereas that of
(grade 3) is seen in young patients, shows aggres- grade I tumour is 7–12%. Hence all patients
sive behaviour with frequent recurrences, brain with grade 2/3 tumours are treated with
invasion and metastasis. Anaplastic (malignant) immediate postoperative radiotherapy.
meningioma is also aggressive.
Data suggest that postoperative radiotherapy
An incidental finding on CT/MRI is one of
improves local control compared with no radio-
the common modes of diagnosis. Presentation
therapy after subtotal resection (15-year local
depends on the location of tumour. Base of skull
control 81% vs. 49%; p = 0.0001).
tumours usually present with cranial nerve
involvement whereas tumours over the convexity Primary CNS lymphoma (PCNSL)
of the cerebrum present with headache or seizure. PCNSL has a higher incidence in immunocompro-
Treatment of meningioma mised individuals than in the normal population.
It commonly presents in the third and fourth
Meningiomas can be observed if they are small
decades in immunocompromised individuals
and asymptomatic. Treatments of choice for pro-
whereas it occurs three decades later in those who
gressive or symptomatic meningioma are either
have a competent immune system. The usual pres-
complete surgical resection if in an operable
entations include focal symptoms, raised intra
location or radical radiotherapy or radiosurgery,
cranial pressure, behaviour and personality
if in an inoperable site. Complete resection is
changes and confusion. Biopsy shows diffuse
usually difficult for skull base (Figure 16.6A),
large cell B cell lymphoma in 80–90%. Up to
cavernous sinus and cerebellopontine angle men-
20–40% patients have CSF involvement. Less
ingiomas. After incomplete resection, either a
than 5% people have systemic involvement.
policy of imaging surveillance with delayed
Tissue diagnosis is made from an open or stere-
radiotherapy on progression or immediate post-
otactic biopsy. Staging investigations include HIV
operative radiotherapy is adopted. The policy of
testing, chest X-ray, CSF examination and slit
image surveillance with delayed radiotherapy
lamp examination (10–20% have uveitis). Sys-
depends on:
temic staging is only indicated if B symptoms are
1. Location of tumour – tumour near critical present (see p. 306).
structures may cause neurological damage The role of surgery is limited to biopsy.
when it progresses and hence early radiother- Steroids may cause complete disappearance of
276
CENTRAL NERVOUS SYSTEM TUMOURS 16
A B
Figure 16.6
Tumours of the base of the brain.
A, Homogeneously enhancing
meningioma. B, Heterogeneously
enhancing craniopharyngioma. C, Cystic
enhancing pituitary adenoma.
tumour (ghost tumours) and hence should be optic pathway (visual field defects) and neuro-
avoided until tissue diagnosis. Current standard logical symptoms.
treatment is high-dose intravenous methotrexate Surgical resection is the recommended treat-
with whole brain radiotherapy (40–50 Gy) which ment if feasible. Following incomplete resection
results in a 2-year survival of 43–73%. However or cyst decompression and biopsy, postoperative
this treatment is extremely toxic particularly in radiotherapy is indicated which results in a
the elderly when 60–80% patients older than 5-year survival of 89% and 10-year survival of
60 years develop progressive leucoencephalopa- 77%.
thy and cognitive dysfunction. Hence in the
elderly chemotherapy alone with delayed radio- Pituitary adenoma and carcinoma
therapy is an alternative. In immunocompro-
mised patients reduction of radiotherapy dose Pituitary adenoma
may be necessary to reduce toxicity. Pituitary adenoma (Figure 16.6C) usually
presents between the ages of 30–50 years. These
Craniopharyngioma are slow growing tumours with an insidious
Craniopharyngioma (Figure 16.6B) arises from onset of symptoms. The usual presentation
the epithelial remnants of Rathke’s pouch. These is hormonal dysfunction or symptoms result-
are slow growing tumours often with a solid and ing from pressure effects such as visual field
cystic component, the latter filled with a fluid defects.
having a ‘motor oil’ appearance. Medical management is important in secre-
The clinical presentation is with pituitary hor- tory tumours. Transphenoidal surgery is the
monal abnormalities, pressure symptoms on the standard surgery for secretory tumours other
277
II SITE-SPECIFIC CANCER MANAGEMENT
A B
C D
Figure 16.7
Various appearances of brain metastases. A, Bleeding metastasis (melanoma). B, Single cystic metastasis (mucinous
carcinoma, breast). C, Multiple metastasis (lung cancer). D, Predominant posterior fossa metastases (HER2+ breast
cancer).
There is no role for surgery in multiple metas- Box 16.5: Single or solitary metastasis
tases, except to relieve hydrocephalus. Patients
• Single metastasis – one lesion within the brain
with highly chemosensitive tumours (e.g. germ irrespective of status of systemic disease
cell tumours, choriocarcinoma) are treated with • Solitary metastasis – the one lesion in the brain is
initial chemotherapy followed by consolidation the only metastatic disease in the body
whole brain radiotherapy in some cases. • Oligometastases – solitary or small number (<4)
metastases which can be treated aggressively
Single or solitary metastasis (on MRI) (Box 16.5)
In a group of patients with operable single or
solitary metastasis, surgical excision followed by previous 3 months. Complete excision followed
WBRT improve survival compared with WBRT by close imaging follow-up may be appropriate
alone (40 weeks vs. 15 weeks; p < 0.01). Most for some patients with good prognosis.
benefit is seen in patients <65 years with KPS Patients with inoperable tumours due to
of ≥70 and no extracranial progression in the inaccessible locations may be considered for
279
II SITE-SPECIFIC CANCER MANAGEMENT
stereotactic radiosurgery followed by WBRT if weeks, with prompt treatment median survival is
they have a PS of 0–1, >1year disease-free inter- around 6–8 months.
val and controlled systemic disease.
Spinal cord tumours
Brain metastasis after prophylactic cranial Spinal cord tumours can be either primary or
secondary. Primary spinal cord tumours consist
radiotherapy and previous treatment
of 2–4% of all primary CNS tumours. Common
Patients with isolated metastasis can be con spinal cord tumours are shown in Box 16.6.
sidered for radiosurgery in a selected group of Pain is the commonest presenting symptom.
patients. If a patient had previously benefited There will be varying degrees of spinal cord com-
from WBRT for at least 4 months and the general pression depending on the location of tumour.
condition is good (PS 0–1), re-irradiation, either Evaluation includes a full neurological examina-
partial brain or whole brain may be considered. tion to assess the extent and level of neurological
Leptomeningeal carcinomatosis deficit. MRI is the imaging of choice. Intramedul-
lary tumours may expand the cord and other
Infiltration of the leptomeninges by cancer com-
imaging features are similar to that of corre-
monly occurs with breast and lung cancer,
sponding tumours in the brain (Table 16.1).
melanoma, lymphoma and leukaemias. Clinical
presentation can be with features of increased Management
intracranial pressure, cranial nerve palsies or General management includes pain control, ster-
impaired higher function. Diagnosis is based on oids which reduce both pain and the neurological
MRI scan (CT has a reduced sensitivity of 30% deficit and rehabilitative measures.
compared with 70% for MRI) and CSF examina- The definitive treatment is surgery. Early
tion. Typical findings on MRI are subarachnoid surgery is advocated to avoid further neurologi-
or parenchymal nodule (Figure 16.8) and sulcal/ cal damage. Maximal safe removal with the help
dural enhancement. Patients with good prognosis of operative microscope and tools such as intra-
(limited disease, PS0–1, chemo-sensitive tumour operative ultrasound and cavitating ultrasonic
and minimal neurologic deficit) are treated with aspirator is the norm. Indication for and dose of
intrathecal chemotherapy with radiotherapy to radiotherapy is as in Box 16.7. Radiotherapy is
symptomatic sites and systemic chemotherapy. indicated for all intramedullary tumours except
Median survival in untreated patients is 4–6 the following situations:
1. Pilocytic tumours (grade 1) after complete and
incomplete resection (5-year survival 90%).
2. Myxopapillary ependymoma (grade 1) after
complete (10-year survival 98%) or incom-
plete resection (10-year survival 70%).
281
Endocrine tumours
TV Ajithkumar
17
Thyroid cancer noma and hemithyroidectomy is indicated for all
follicular lesions. MRI is the preferred imaging
Thyroid malignancies are the commonest endo- to assess local invasion (Figure 17.1) as the use
crine malignancy. Common types of thyroid of iodine containing contrast can cause thyroid
cancers are papillary and follicular carcinomas stunning which may prevent effective use of radi-
(well-differentiated cancers) arising from epithe- oiodine for at least three months subsequently.
lial cell, medullary carcinomas from the para
follicular C cells, anaplastic carcinoma and Staging
NHL. Well-differentiated cancers typically affect Box 17.1 shows the TNM staging system. The
women in their 40s. The median age of presenta- TNM and MACIS (Box 17.2) have been shown
tion of medullary carcinoma is 50–60 years and to be the best predictors of outcome in differenti-
older for anaplastic carcinoma. ated thyroid cancer.
Aetiology Management
The majority of thyroid cancers are sporadic. Well-differentiated cancer
Other risk factors include radiation exposure Surgery is the definitive treatment of all malig-
(papillary carcinoma), genetic (e.g. MEN2 nancies except lymphoma. The majority of
with medullary carcinoma, FAP with papillary patients need total or near total thyroidectomy.
carcinoma) and endemic goitre (follicular In low-risk patients (<1 cm papillary cancer with
carcinoma). no nodes, <2 cm follicular cancer in women aged
less than 45 years and <1 cm follicular cancer
Presentation with minimal capsular invasion) lobectomy is
Common presentation is with an enlarging adequate. Patients with high-risk disease (male
solitary nodule. Cervical lymphadenopathy is >45 years, tumour >4 cm and extracapsular
common with medullary, papillary and anaplas- spread) are at risk of level VI nodal involvement
tic carcinoma. Local invasion can lead to symp- and hence elective central node dissection is indi-
toms of hoarseness of the voice and dysphagia. cated. Patients with proven neck node metastases
Anaplastic carcinoma has an aggressive growth are treated with modified radical neck dissection.
with local invasion and rapid growth which can All patients should have serum calcium postop-
lead to stridor. eratively to rule out transient hypocalcaemia.
Serum thyroglobulin (TG), the tumour marker,
Evaluation is estimated 6 weeks postoperatively which
Evaluation includes clinical examination and should be undetectable in patients who had total
assessment for thyrotoxicosis which is rare in thyroidectomy and have no residual disease.
cancer. Initial investigations include thyroid Patients after total thyroidectomy are started on
function tests and FNA of the lesion. Biopsy may triiodothyronine (T3) 20 mg thrice daily which
be indicated for lymphoma and anaplastic carci- is subsequently changed to T4. T4 is started
282 © 2011, Elsevier Ltd
DOI: 10.1016/B978-0-7234-3458-0.00022-1
ENDOCRINE TUMOURS 17
A B
Figure 17.1
Axial (A) and coronal (B) view of MRI scan in anaplastic carcinoma of thyroid.
Box 17.1: TNM staging in thyroid cancer Box 17.2: MACIS (Metastasis, Age, Completeness
of resection, Invasion, Size). Scoring system to
T stage
assess 20-year survival probability of papillary
pT1 Tumour ≤2 cm in greatest dimension
pT2 Intrathyroidal tumour >2–4 cm in greatest Scores
dimension Metastasis – Present: score 3
pT3 Intrathyroidal tumour >4 cm in greatest Age – <40 years: score 3.1
dimension >40 years: score = 0.08 × age
pT4 Tumour beyond the thyroid capsule
Completeness of resection – incomplete: score 1
N stage (cervical and upper mediastinal)
Invasion – present: score 1
N0 No nodes involved Tumour size: score = 0.3 × size in cm
N1 Regional nodal involvement
N1a Ipsilateral cervical nodes Total score and 20-year survival
N1b Bilateral, midline or contralateral cervical
<6 99%
nodes, or mediastinal nodes 6–6.99 89%
Distant metastases 7–7.99 56%
>8 24%
M0 No distant metastases
M1 Distant metastases
MX Distant metastases cannot be assessed
Staging of epithelial thyroid carcinomas with a dose of 100 mcg daily and increased by
Stage I 25 mcg every 6 weeks until TSH levels are below
Aged 45 or younger with any pT, any N M0 0.1 MiU/L.
Aged 45 or older with pT1N0M0
Radioablation
Stage II
Aged 45 or younger with any pT any N M1
Postoperative radioiodine ablation is recom-
Aged 45 or older with pT2N0M0 or pT3N0M0
mended for all patients except for those who had
total thyroidectomy for a well-differentiated uni-
Stage III
Aged 45 or older with pT4N0M0 or any pT, N1M0
focal tumour of <1 cm with no vascular or extra
capsular invasion. Informed consent should be
Stage IV
Aged 45 or older with any pT, any N, M1 obtained and instruct female patients not to get
Anaplastic thyroid carcinoma any age or stage pregnant prior to and up to 12 months after
treatment (Box 17.3). T4 is substituted with T3
4–6 weeks before radioablation and T3 is with-
drawn 2 weeks before ablation. 3–3.7 GBq of
radioiodine is administered orally. In patients
with a history of myxoedema psychosis, severe
283
II SITE-SPECIFIC CANCER MANAGEMENT
Early effects
• Sialoadenitis and taste changes • Start T3 20 mg TDS initially and then change to T4
• Nausea • Serum thyroglobulin at 6 weeks
• Neck pain and swelling
• Radiation cystitis, gastritis • Change T4 to T3 4–6 weeks prior to radioblation
• Stop T3 2 weeks prior to radioblation
• Symptoms of hypothyroidism for 4–6 weeks
Late effects
• Dry mouth and taste changes Oral radioiodine 3–3.7 GBq
• Dry eyes due to lachrymal gland dysfunction
• Increase in miscarriage rate in first year after Uptake scan 3–10 days later and serum thyroglobulin
treatment
• Pulmonary fibrosis
• Male infertility If uptake or raised thyroglobulin
No uptake or
• Risk of 2nd malignancy raised TG
Surgery or 5.5 MBq radioiodine
cardiac disease and high volume disease, T3 Stimulated TG estimation and radioiodine uptake scan at 6 months
withdrawal can be dangerous and hence recom-
binant human TSH (0.9 mg IM daily for 48 Increased TG and Increased uptake
hours before radioablation) is used. Patients are no uptake on scan on scan
admitted to and remain in a specialist facility
with appropriate shielding and sanitation. Isola- Repeat stimulated TG Surgery if feasible or
tion is continued until safe levels of radiation are at 6 months radioiodine 5.5 MBq
measured by a medical physicist. Patients are
advised to keep well-hydrated, empty the bladder Decreasing Increasing Uptake scan at 3-20 days
frequently and to take prescribed laxatives to after radioiodine
avoid constipation and these measures are aimed
Follow-up PET scan
at minimizing the absorbed dose of radiation. An 6 monthly radioiodine
uptake scan is done 3–10 days after radioiodine 5.5 MBq until no more
uptake or bone marrow
to assess residual thyroid tissue. In patients with suppression or lung
elevated TG and positive uptake scans, either No disease Disease present toxicity
further surgery or repeat dose of radioiodine
(5.5 MBq) is indicated. Further management Observation or
afterwards is shown in Figure 17.2. empirical radioiodine
Some patients do not have a take-up of radio- Figure 17.2
iodine when external beam radiotherapy to the Management of well-differentiated thyroid cancer.
entire thyroid bed has been carried out, and level
III–VI nodes (60 Gy in 30 fractions) is indicated
if there is gross residual disease, extranodal
disease and in patients >60 years with T4 disease. if metastatic disease is present. In metastatic
disease this helps with optimal local symptom
Medullary carcinoma control and control of calcium levels. In patients
All patients with medullary carcinoma should with level II–V nodal involvement modified
have screening for phaeochromocytoma with radical neck dissection is done. All patients need
24-hour urinary catecholamines and serum a suppressive dose of thyroxine.
calcium to exclude hyperparathyroidism (MEN2 There is no role for radioiodine ablation. In
syndrome). patients with incomplete resection, postoperative
Total thyroidectomy with elective central radiotherapy to the thyroid bed, bilateral cervical
nodal dissection is the treatment of choice even and upper mediastinal nodes (50–60 Gy in 2 Gy
284
ENDOCRINE TUMOURS 17
per fraction) may be given to improve relapse- virilization is the most common presentation.
free survival. Other presenting symptoms include virilization in
Follow-up is life-long with clinical examina- women and gynaecomastia and testicular atrophy
tion, serum calcitonin estimation and ensuring in men, hypertension and hypokalaemia.
adequate thyroid suppression. Non-secreting tumours present with abdomi-
Patients presenting with recurrence may be nal discomfort or back pain due to mass effect.
treated with surgical excision if feasible. Radio- Rarely patients can present with fever, weight
therapy is useful in inoperable local recurrence. loss and anorexia.
Patients with symptomatic metastatic disease
may be considered for 131I-MIBG or radiolabelled Diagnosis
somatostatin analogues if there is uptake with Initial investigations include hormonal workup
the corresponding scans. and imaging with CT chest and abdomen (Figure
All patients with medullary carcinoma thyroid 17.3). MRI is also useful in identifying invasion
require genetic screening for mutations in the of adjacent organs and the inferior vena cava,
RET gene at exon 10, 11, 13 and 16. Prophylac- and hence in planning surgery. Biopsy of an
tic surgery is indicated in gene carrier mutated adrenal tumour is controversial because of the
relatives (before 1 year in MEN 2B and 5–10 theoretical risk of needle tract metastases.
years in MEN 2A). However, it may be acceptable if primary surgical
management is not feasible and diagnosis cannot
Anaplastic carcinoma thyroid be established with non-invasive measures.
Surgery is an option only in a small proportion
of patients and the majority of patients present Staging
with locally advanced and metastatic disease. WHO (2004) staging for adrenocortical carci-
Radiotherapy is the principal treatment which is noma is as follows:
essentially to improve local control. It is given
Stage I: localized tumour of ≤5 cm.
either as 50–60 Gy in 2 Gy per fraction or in
Stage II: localized tumour of >5 cm.
combination with chemotherapy (adriamycin).
Prognosis
The long-term survival of well-differentiated
thyroid cancer is >90%. 10-year survival of med-
ullary carcinoma thyroid is 50–70% and median
survival of anaplastic carcinoma is less than 6
months (5-year survival <10%).
Adrenocortical carcinoma
Introduction
Adrenocortical carcinoma (ACC) is a rare malig-
nancy with heterogeneous manifestations and a
poor prognosis. In adults it peaks at 57 years of
age. There are no proven aetiological factors.
ACC is rarely reported in multiple endocrine
neoplasia I (MEN I), Li–Fraumeni and Wiede-
mann–Beckwith syndromes.
Figure 17.3
Presentation CT scan in adrenocortical carcinoma. On CT scan
adrenocortical carcinoma is typically more than 4 cm
Approximately 60% patients present with fea- with central necrosis with heterogeneous enhancement
tures of adrenal steroid hypersecretion. Rapidly and may show calcification (30%) and invasion into
progressing Cushing’s syndrome with or without adjacent structures.
285
II SITE-SPECIFIC CANCER MANAGEMENT
286
Cancers of
unknown primary
TV Ajithkumar and HM Hatcher
18
Introduction entity which may be a favourable (15% patients)
or unfavourable (85% patients) (Box 18.1).
Cancers of unknown primary (CUP) constitute All patients should have an assessment of
3–5% of all cancers. The criteria for diagnosis of performance status and co-morbidities, which
CUP includes patients who have a biopsy proven will guide treatment decisions.
metastatic cancer in whom a detailed history,
physical examination, blood tests, immunohisto- Management
chemistry of biopsy, chest X-ray, CT scan of the
abdomen and pelvis and in certain cases, mam- The decision to treat depends on the clinico-
mography or FDG-PET fail to detect a primary pathological entity and performance status.
tumour. Patients with poor performance status (>2) are
generally treated symptomatically. Treatment of
Evaluation various subsets is as described below.
Clinicopathological evaluation aims to identify a Women with isolated axillary
primary site, a clinicopathological subset and to lymphadenopathy
rule out non-epithelial tumours. The first step Women presenting like this are treated like stage
includes clinical history to establish risk factors, II breast cancer. Previous studies showed an
clinical examination, investigations and patho- occult primary in 50–60% of cases after mastec-
logical evaluation. Initial investigations include tomy. However, if MRI of the breast failed to
blood tests, urinalysis, faecal occult blood test and show any primary, breast surgery or radiother-
CT scan of the chest, abdomen and thorax. Endo- apy may not be warranted. Axillary lymphaden-
scopic evaluation is guided by symptoms and/or opathy is treated with lymph-node dissection
signs. In patients suspected to have extragonadal followed by regional radiotherapy based on
germ cell tumours a serum beta-hCG and AFP breast cancer radiotherapy principles (p. 124).
should be estimated. Other tumour markers Adjuvant systemic treatment is similar to stage II
depending on the clinical context include PSA, breast cancer. The reported 5-year survival for
CEA, CA-125 and CA19-9. Pathological evalua- patients with 1–3 nodes are 87% and those with
tion includes light microscopy and expedient use >3 nodes are 42%.
of immunohistochemical techniques. Immunohis-
tochemistry is useful in differentiating epithelial Women with peritoneal carcinomatosis
from non-epithelial tumours, such as sarcoma and Women with peritoneal carcinomatosis are
lymphoma (Table 18.1). The majority of epithe- treated like stage III ovarian cancer with initial
lial tumours are adenocarcinoma (65–80%) fol- cytoreduction followed by combination chemo-
lowed by squamous cell carcinoma (10–15%) and therapy or vice versa depending on performance
undifferentiated carcinoma (5–10%). status and co-morbidities (p. 201). CA-125 may
The outcome of first step evaluation is be used as a tumour marker if elevated and the
identification of a specific clinicopathological reported long-term survival is 15%.
*Adenocarcinoma in lung – TTF-1 is highly specific for lung and thyroid adenocarcinoma, lung small cell, and may stain extra-pulmonary
small cell carcinoma.
289
Cancer in pregnancy
TV Ajithkumar and HM Hatcher
19
Cancer during pregnancy poses a very challeng- been suggested due to the risk of foetal heating/
ing situation. Cancer complicates 0.02–0.1% of cavitation. Abdominal X-rays, CT scans and
pregnancies. The oncologist faces the difficult isotope scans should be avoided during preg-
situation of optimizing the care of the mother nancy. However, a final decision to perform
without causing harm to the foetus. The common these investigations is based on the risk benefit
tumours diagnosed during pregnancy are malig- for the mother (your patient), their expected
nant melanoma, breast cancer and cervical cancer. survival and the gestation of the foetus.
Box 19.1: Uterine/foetal radiation dose during Box 19.2: Foetal radiation dose
various radiological investigations during radiotherapy
Investigation Uterine/foetal dose Radiotherapy Foetal dose
Chest X-ray 0.000005 Gy Cervical cancer 45–50 Gy
Abdominal X-ray 0.022 Gy Mantle field 0.014–0.13 Gy
Mammogram 0.04 Gy Breast cancer 0.14–0.18 Gy
Chest CT scan 0.002 Gy Brain tumour and head and neck 0.0015–0.08 Gy
Abdominal CT scan 0.02 Gy cancer
Pelvic CT 0.07 Gy
Barium enema 0.036 Gy
IVU 0.045 Gy
Bone scan 0.018–0.045 Gy than 100 mGy (Box 19.2). During radiotherapy
1 Gy = 100 cGy = 1000 mGy to the breast and chest wall, the dose to the
foetus increases with gestational age. The foetal
dose depends on the radiation field, dose, the
hypotension, and long-term supine positioning distance between the edge of radiation field
of mother during surgery causing placental and foetus, and the machine and its shielding
underperfusion. measures.
In the third trimester surgery after foetal matu- Radiotherapy should be delayed until postpar-
ration and delayed surgery until post-partum if tum if possible. If radiotherapy cannot be post-
appropriate are the options. poned the second trimester is the preferable time
Intra-abdominal surgery is more likely to inter- to deliver with appropriate precautions to keep
fere with pregnancy. During major abdominal foetal dose below 5–10 cGy.
and pelvic surgery, continuous monitoring of the
foetus is necessary. Measures to suppress prema- Systemic treatment
ture labour and prophylactic treatment to improve Most cytotoxic drugs have a low molecular
foetal lung maturity should be undertaken if the weight of less than 600 kDa and can cross the
surgery carries a risk of premature labour. placenta. The concern is that these agents can
cause various mutagenic, teratogenic and carci-
Timing of delivery and nogenic effects in the foetus. Physiological
breast feeding changes due to pregnancy such as change in
haemodynamics and renal clearance will also
Delivery is generally planned after weeks 32–35
alter the metabolism, distribution and elimina-
(earlier in modern neonatal units with better
tion of cytotoxic agents leading to unpredictable
outcome for the baby) and 3 weeks after the last
effects. Chemotherapy interferes with organo-
chemotherapy. Breast feeding is not recom-
genesis in the first trimester, leading to a high risk
mended during chemotherapy, up to 2–4 weeks
of teratogenesis (10% with single agent and 20%
after completion of chemotherapy and during
with combination) and hence is not advised.
hormonal treatment.
After the first trimester organogenesis is complete
except for the brain and gonads. Chemotherapy
Radiotherapy during this period can cause foetal growth retar-
The developing embryo or foetus is extremely dation (2-fold risk), stillbirths (5%), premature
sensitive to ionizing radiation, which can cause delivery (5%), low birth weight (7%) and myelo-
foetal loss, malformations, growth retardation suppression (4%). However, the risk of tera-
and defects in intelligence. Radiotherapy is there- togenesis is almost similar to the general
fore contraindicated for abdominal and pelvic population (2–3%); but there is concern about
malignancies during pregnancy. Several studies risk of sterility, defects in intellectual function
support the use of radiotherapy for head and and other organ dysfunction due to in utero
neck cancer, breast cancer and brain tumours. exposure of chemotherapy.
Radical radiotherapy to the head and neck and Alkylators and anti-metabolites are the most
brain can be achieved with a foetal dose of less teratogenic and have high abortive potential.
291
II SITE-SPECIFIC CANCER MANAGEMENT
Psychosocial support
A diagnosis of cancer during pregnancy is an
extremely distressing situation for the patient and
family, as they may have to decide about future
plans which may not include both of them. This
necessitates provision of adequate psychosocial
support for both the parents (and probably grand-
parents!). Needless to say that there will be anxiety
about the baby, though evidence shows little harm
to date (but is minimal). The risks of a genetically
associated cancer in the child should be evaluated
Figure 19.1
Pregnancy associated ovarian cancer – MRI shows uterus by clinical genetics criteria (p. 45) but are not
with foetus (white arrow heads) and omental thickening affected by being diagnosed in pregnancy.
(arrow). Biopsy of the omental thickening confirmed
serous papillary adenocarcinoma consistent with a
primary peritoneal cancer. Further reading
Azim HA Jr, Peccatori FA, Pavlidis N. Treatment of the
pregnant mother with cancer: a systematic review on the
use of cytotoxic, endocrine, targeted agents and
stromal tumour (most common dysgerminoma). immunotherapy during pregnancy. Part I: Solid tumors.
Cancer Treat Rev. 2010;36:101–109.
Most of the borderline and germ cell tumours are
Azim HA Jr, Pavlidis N, Peccatori FA. Treatment of the
diagnosed at an early stage. pregnant mother with cancer: a systematic review on the
Staging includes chest X-ray, abdominal and use of cytotoxic, endocrine, targeted agents and
pelvic ultrasound and MRI if indicated (Figure immunotherapy during pregnancy. Part II: Hematological
19.1). CA-125, beta-hCG and AFP are raised in tumors. Cancer Treat Rev. 2010;36:110–121.
Kal HB, Struikmans H. Radiotherapy during pregnancy: fact
pregnancy.
and fiction. Lancet Oncol. 2005;6:328–333.
Laparotomy or laparoscopy is indicated when Pentheroudakis G, Pavlidis N. Cancer and pregnancy:
there is suspicion of an ovarian lesion (persistent poena magna, not anymore. Eur J Cancer. 2006;42:
solid or complex mass lesion of >6 cm with asso- 126–140.
294
20
Human
immunodeficiency
virus (HIV) related
malignancies
TV Ajithkumar
Box 20.1: Cancers in HIV patients Box 20.2: Staging of Kaposi’s sarcoma
AIDS-defining cancers Good risk (all of Poor risk (any of the
• Kaposi’s sarcoma the following) following)
• Invasive cervical cancer Tumour (T) Confined to skin • Associated oedema
and/or lymph or ulceration
• Non-Hodgkin’s lymphoma
nodes and/or • Oral disease other
• Burkitt’s non-nodular than non-nodular
• Immunoblastic disease in disease in palate
• Primary cerebral palate
• Gastrointestinal KS
Non-AIDS-defining cancers • Non-nodal visceral
• Anal cancer KS
Systemic No history of History of
• Liver cancer illness opportunistic opportunistic
• Hodgkin’s disease infection or infection or thrush
• Penile cancer thrush B symptoms present
• Vulva and vagina cancer No B symptoms* <70 KPS
• Oral cavity and pharynx ≥70 KPS Other HIV-related
illness
• Larynx
*B symptoms – unexplained fever, night sweats, >10% involuntary
• Lung
weight loss or diarrhoea persisting more than 2 weeks
• Myeloma
• Acute myeloid and monocytic leukaemia
Liver cancer
Hepatocellular cancer is increased by 8-fold in
Optimal treatment of HIV-related lymphoma
the HIV population. Tumours tend to be more
is not known. Patients are generally treated simi-
symptomatic, advanced at presentation and have
larly to those who are seronegative (p. 302). The
an aggressive course. Resection or transplanta-
overall prognosis is poor with a less than 1 year
tion is an option for early stage whereas systemic
median survival.
chemotherapy is used in advanced cancer (p.
Primary CNS lymphoma is 15 times more
147). Survival is not influenced by CD4 count.
common than in the general population. The
histology is large cell or immunoblastic type of B
Hodgkin’s disease
cell origin. Treatment involves radiotherapy com-
bined with steroids, which extend the median Patients present early in HIV infection with
survival from 2–3 months to 6–8 months (p. 306). unfavourable histologic subtypes (mixed cellu
larity, lymphocyte depleted). There is a 5–15 fold
Non-AIDS-defining malignancies increase in risk with a high rate of EBV co-
infection (75–100%). Clinical presentation with
These account for the increased mortality in the B symptoms and extranodal disease are common.
HAART era. HIV-infected people have an They are treated as seronegative patients (p. 298)
increased risk of these cancers (RR 1.9), and and the median survival is 12–18 months.
these tumours occur at a relatively younger age.
Apart from duration of immunosuppression, Other cancers
other contributing risk factors include HAART Other cancers are treated similarly to seronega-
interruption, smoking, sun exposure, oncogenic tive patients.
viruses and family history. The diagnosis needs
to be confirmed by biopsy and staging will be
affected by the presence of reactive lymphaden- Interactions between
opathy as well as unrelated imaging abnormali- chemotherapy and HAART
ties. Treatment is based on performance status,
There are interactions between some cytotoxic
co-morbidity and potential for surgery. Non-
agents and antivirals due to cytochrome p450
medical management is also challenging due to
enzymes and p-glycoprotein. These interactions
chemotherapy-enhanced immunosuppression,
may account for the better response rate and
added cytotoxicity, drug interaction with
higher rates of survival seen with combinations
HAART and severe radiation reactions. Patients
of cytotoxics and antivirals compared with cyto-
need regular monitoring of their CD4 count.
toxics alone in patients with HIV associated
Continuation of HAART, prophylaxis of oppor-
malignancies. These interactions can also result
tunistic infections and supportive medications
in higher toxicities with treatment.
such as G-CSF are important.
Anal cancer Further reading
The high-risk group includes patients who prac- Spano JP, Costagliola D, Katlama C et al. AIDS-related
tice receptive anal intercourse, men who have sex malignancies: state of the art and therapeutic challenges.
J Clin Oncol. 2008;26:4834–4842.
with men and anal co-infection with HPV or
Deeken JF, Pantanowitz L, Dezube BJ. Targeted therapies to
syphilis. The clinical presentation and treatment treat non-AIDS-defining cancers in patients with HIV on
is similar to that of the seronegative population HAART therapy: treatment considerations and research
(p. 170). Patients with high-grade anal intra- outlook. Curr Opin Oncol. 2009;21:445–454.
297
Cancers of the
haematopoietic system
TV Ajithkumar and HM Hatcher
21
Hodgkin’s disease cells in a background of non-neoplastic cells such
as lymphocytes, histiocytes, neutrophils, eosi-
Epidemiology nophils and monocytes. Classical HD is charac-
terized by CD30 positive H-RS cells and the
Around 1500 cases of Hodgkin’s disease are nodular lymphocyte predominant form of HL
diagnosed every year in the UK and approxi- (LPHL) with CD20 positive lymphocytic and his-
mately 350 people die of this disease every year. tiocytic (L and H) cells. In classical HD, RS cells
Males are predominantly affected (male : female stain positive for CD15 (80%) and CD30 (90%).
1.8 : 1). There is bimodal peak of incidence with
the first peak in those between 15 and 30 and the Clinical features
second peak between 50 and 60 years. The most common presentation is lymphaden-
opathy in the cervical or supraclavicular region
Aetiology (60–70%). Almost two-thirds of patients with
• EBV increases the risk of HD by two- to classical HL have mediastinal nodal disease pro-
threefold. In up to 50–90% of classical HD, ducing symptoms: tight chest, cough, venous
RS cells stain for EBV DNA. congestion, or dyspnoea. About 40% of patients
• Family members of patients affected by HL present with ‘B-symptoms’ of fever >38°C,
are at a three to nine fold increased risk of drenching night sweats, or weight loss >10%
developing the same disease. Same sex sib- within the previous 6 months (not from other
lings have 10 times higher risk of HD. causes). Bone marrow involvement at presenta-
tion occurs in less than 10% of cases. Extranodal
Pathology involvement commonly occurs in the lung.
The WHO classification of HD is shown in Table Investigations and staging
21.1. Nodular sclerosis is the common subtype • Blood – full blood count and ESR, biochem-
in young adults (more common in females), istry including LDH.
which presents with early stage supradiaphragm • Tissue diagnosis – An excision biopsy of a
atic disease whereas mixed cellularity presents lymph node is required for accurate diagnosis
with generalized lymphadenopathy or extra and subtyping. All patients need bone marrow
nodal disease with B symptoms. Lymphocyte examination.
depletion type presents with advanced stage
disease with extranodal involvement and aggres- Imaging
sive clinical course. • Chest X-ray may show mediastinal widening
LRCHL usually presents in young males with (Figure 21.1), pleural effusion or pericardial
localized cervical node involvement with no B effusion.
symptoms. • CT of the neck, thorax, abdomen and pelvis
Histologically HD is characterized by the will delineate lymphadenopathy as well as
presence of Hodgkin and Reed–Sternberg (H-RS) visceral involvement.
298 © 2011, Elsevier Ltd
DOI: 10.1016/B978-0-7234-3458-0.00026-9
CANCERS OF THE HAEMATOPOIETIC SYSTEM 21
Table 21.1: WHO classification of Hodgkin’s Table 21.2: The Cotswolds modification of
lymphoma Ann Arbor staging for lymphoma
Classical Hodgkin’s lymphoma Frequency Stage I Involvement of a single lymph node
region or lymphoid structure or
Nodular sclerosis Hodgkin’s 60–70%
involvement of a single
lymphoma (grades 1 and 2)
extralymphatic site (IE)
Mixed cellularity Hodgkin’s lymphoma 20–30%
Stage II Involvement of two or more lymph
Lymphocyte-rich classical Hodgkin’s 3–5% node regions on the same side of the
lymphoma (LRCHL) diaphragm; localized contiguous
involvement of only one extranodal
Lymphocyte-depleted Hodgkin’s 0.8–1%
organ or site and lymph node
lymphoma (LDHL)
region(s) on the same side of the
Nodular lymphocyte-predominant 3–5% diaphragm (IIE)
Hodgkin’s lymphoma (LPHL)
Stage III Involvement of lymph node regions
on both sides of the diaphragm
(III),which may also be accompanied
by involvement of the spleen (IIIS) or
by localized contiguous involvement
of only one extranodal organ site
(IIIE) or both (IIISE)
Stage IV Diffuse or disseminated involvement
of one or more extranodal organs or
tissues, with or without associated
lymph node involvement
Designations applicable to any disease stage
A: No B symptoms
B: Presence of B symptoms
X: Bulky disease (a widening of the
mediastinum by more than one third
of the chest or presence of a nodal
mass with a maximal dimension
greater than 10 cm)
Figure 21.1
Mediastinal widening in Hodgkin’s disease.
g
Survival
h i
With combined modality treatment patients in
the early-favourable group achieve a disease free
survival (DFS) of more than 90% and an overall
survival of about 95% at five years. Patients in
j k the early unfavourable group have a DFS of
about 84% and an OS of 91%. In advanced stage
combination chemotherapy results in 5-year
failure free survival of 78% and overall survival
Figure 21.2
Nodal regions: cervical (a-b), mediastinal (c), axillary
of 90%. High-dose therapy and autologous stem
(d-e), mesenteric (f), paraaortic (g), iliac (h-i) and cell transplantation results in DFS of 55% and
inguinal (j-k). the overall survival 71% at 5 years.
Advanced stage
Follow-up
Combination chemotherapy with 6–8 cycles of Two-thirds of relapses occur within 3 years of
ABVD is the current treatment of choice; although initial treatment and more than 90% occur
this regime is not as successful as in earlier stage within 5 years. Since a significant proportion of
disease. Various attempts are being made to patients who relapse can be successfully salvaged,
modify the ABVD regime by adding newer drugs all patients need regular follow-up (Box 21.4).
and increasing dose intensity. Follow-up is also helpful in identifying and man-
The role of consolidation radiotherapy in aging long term side effects such as endocrine
advanced stage HD is also not clear. A meta- dysfunction.
analysis of combined modality treatment showed
equal tumour control but inferior overall survival
Survivorship issues
compared with chemotherapy alone. An EORTC The major causes of excess death in survivors of
study suggested that involved field radiotherapy HD are second malignancies and ischaemic heart
did not result in better outcomes in patients who disease (p. 58).
achieve complete response after chemotherapy, One of the most common second malignancies
but may be beneficial in patients with a partial is lung cancer which is attributed to mediastinal
response. Hence consolidation radiotherapy is radiotherapy, chemotherapy and smoking.
considered in those with initial bulky disease and Patients who had thoracic radiotherapy are
those who fail to achieve a complete response to encouraged not to smoke. There is 20–50% risk
chemotherapy. of second breast cancer after mediastinal/axillary
radiotherapy in young females and these patients
Recurrence should be screened for breast cancer. Common
Combination chemotherapy is usually the treat- haematological neoplasms include acute myeloid
ment of choice for patients who relapse after leukaemia, myelodysplastic syndrome (develops
301
II SITE-SPECIFIC CANCER MANAGEMENT
Follicular lymphoma
Extranodal marginal zone B-cell lymphoma of mucosa-associated lymphatic tissue (MALT) type
Burkitt lymphoma
tional prognostic index (IPI) is used for patients disease and median survival is 10–15 years from
with diffuse large B-cell lymphoma (Box 21.6). diagnosis. Spontaneous regression can occur
and 40% of patients transform to an aggressive
Treatment histologic type. Over 90% have rearrangement
Follicular lymphoma of the bcl-2 gene which inhibits apoptosis.
Follicular lymphoma accounts for approximately Early stage
20% of all lymphomas and has a variable clinical Only about one-third of patients with follicular
course. The median age at presentation is 50 lymphoma present with stages I and II or limited
years. Most patients present with advanced stage III (with up to five involved lymph nodes 303
II SITE-SPECIFIC CANCER MANAGEMENT
A B
Figure 21.3
CT scan in NHL. Figure shows massive para-aortic lymphadenopathy (A) and iliac and inguinal lymphadenopathy (B).
A B
Figure 21.4
CT scan in primary CNS lymphoma. Non-contrast enhancing CT scan shows a periventricular hyperdense lesion (A)
which uniformly enhances on contrast administration (B).
306
CANCERS OF THE HAEMATOPOIETIC SYSTEM 21
scrotal swelling. DLBCL is the most common
histology and diagnosis is usually established
by initial orchidectomy. Approximately 80% of
cases are stage I or II at presentation. A low IPI,
no B-symptoms, the use of anthracyclines, and
prophylactic contralateral scrotal radiotherapy
are significantly associated with longer survival.
Systemic chemotherapy alone seems not to be
effective in preventing relapses in the contralat-
eral testis because the testis is a sanctuary site
(p. 35). Treatment consists of 6–8 courses of
R-CHOP followed by prophylactic irradiation
of the contralateral testis. Although data are Figure 21.5
not convincing, prophylactic intrathecal therapy Cutaneous lymphoma. From Darell Rigel: Cancer of the
Skin (Saunders), with permission.
should be considered. This approach results in a
3-year overall survival of 86% and 3-year pro-
gression-free survival 77%.
logic evidence of leukaemic involvement and can
Cutaneous NHL be preceded by mycosis fungoides. The clinical
Primary cutaneous lymphomas differ considera- behaviour of the disease is aggressive.
bly in their clinical course and outcome from Primary cutaneous marginal zone B-cell lym-
systemic lymphomas. Primary cutaneous lym- phoma and primary cutaneous follicle centre
phomas are generally divided into lymphomas B-cell lymphoma usually show an indolent
with an indolent or an aggressive clinical course. clinical behaviour.
Approximately 75% of primary cutaneous lym- Spontaneous resolution may occur. Primary
phomas are T-cell lymphomas which include cutaneous follicle centre B-cell lymphoma prefer-
mycosis fungoides (MF) and Sezary syndrome entially involves the head and trunk with solitary
(SS) and 25% are primary cutaneous B-cell or grouped plaques and tumours. The 5-year sur-
lymphomas. vival is excellent with more than 95% in both
entities.
Clinical features
In contrast, primary cutaneous diffuse-large
Mycosis fungoides typically presents with ery-
B-cell lymphoma, leg-type, behaves aggressively
thematous patches, plaques and tumours usually
and affects predominantly elderly patients. This
in areas not often exposed to sunlight (Figure
entity typically presents as red solitary or multi-
21.5). Most patients have multiple lesions and
ple nodule on the leg but can also rarely be found
ulcerations can occur. The course of the disease
at other sites. Both cutaneous relapses and extra-
is indolent.
cutaneous dissemination are frequent. With a
The lesions of mycosis fungoides can be clas-
5-year survival of only 50%, prognosis is signifi-
sified into four groups: Patches, papules and/or
cantly worse than in the other two entities.
plaques affecting less (T1) or more (T2) than
10% of body surface, T3 with tumours >1 cm Management of cutaneous T-cell lymphoma
and T4 with erythroderma affecting more than In patients with early stages of MF topical
80% of body surface. In T1 disease outcome is therapy with mechlorethamine or bexarotene,
excellent and also patients with T2 disease have superficial radiotherapy and phototherapy
a median overall survival of more than 10 years. (PUVA) are appropriate treatment options.
Patients with erythrodermic mycosis fungoides Patients with more advanced disease will
have a median survival of 5 years and those with require systemic treatment. Total skin electron
visceral involvement have an even worse survival beam therapy (TSEBT) is appropriate in patients
with only one or two years. with generalized thickened plaques due to its
Sezary syndrome is defined as an erythroder- depth of penetration. The rate of complete
mic cutaneous T-cell lymphoma with haemato- responses is greater than 80% but the long-term
307
II SITE-SPECIFIC CANCER MANAGEMENT
outcome is not affected. TSEBT should be previously metabolically active area becomes
followed by an adjuvant therapy such as inactive after treatment.
mechlorethamine or PUVA. Several new monoclonal antibodies are under
Systemic therapy with the orally administered investigation including atumumab (a human
retinoid bexarotene can achieve response rates of monoclonal IgG1 antibody against a small loop
50%. MF and SS are relatively chemoresistant epitope on the CD20 molecule) and epratuzumab
diseases. The most frequently used combination (anti-CD22). Radioimmunotherapy (RIT) is an
chemotherapy regimens include cyclophospha- innovative treatment modality that combines the
mide, vincristine and prednisone (CVP) with or tumour cell targeting ability of monoclonal anti-
without doxorubicin. High-dose chemotherapy bodies with the cytotoxic effect of radiation by
followed by autologous stem cell transplantation linking a radioisotope to the antibody. This is
in patients with advanced disease has been shown under investigation for NHL with 90Y-ibritu-
to induce high response rates in most patients momab tiuxetan and 131I-tositumumab. Other
but the responses were predominantly of short agents under investigation include bortezomib
duration. In contrast, allogeneic transplantation (30–40% response) and lenalidomide.
seems to induce long-term durable remissions of
more than 3 years. Follow-up in NHL
The purpose of follow-up is to detect early
Management of cutaneous B-cell lymphoma
relapse and to educate about possible long-term
Due to their indolent course and excellent
complications. A typical schedule is 3-monthly
outcome in primary cutaneous marginal zone
follow up for 1 year, 4-monthly during the 2nd
and follicle centre B-cell lymphoma a watch-and-
year, 6 monthly for the 3rd year, annually up to
wait strategy is the adequate management in
10 years and then on alternate years. At each visit
most cases. In patients with limited symptomatic
detailed history is taken and clinical examination
skin lesions, local excision or radiotherapy (20–
is done with routine bloods including LDH and
36 Gy) are the first treatments of choice. Cutane-
ESR. Chest X-ray is done in patients with chest
ous relapses can be treated in the same way as
disease at presentation. Further investigations are
the initial lesion and do not worsen prognosis.
needed depending on any new symptoms or signs
In patients with extensive skin lesions rituxi-
or other abnormal tests.
mab is the treatment of choice. Oral chloram-
bucil is a treatment option often used in Europe.
A multiagent chemotherapy is rarely indicated in Acute leukaemias
these types of cutaneous lymphomas with the
exception of patients developing extracutaneous Introduction
disease. Acute lymphoblastic leukaemia (ALL) occurs
most frequently either between the ages of 15–25
The role of stem cell transplantation
(p. 321) or over the age of 75. In the UK there
in aggressive lymphoma are 200 new adult cases per year and the
Autologous stem cell transplantation has become male : female ratio is roughly equal. Acute myeloid
the standard treatment in younger patients with leukaemia (AML) occurs more frequently and the
a first chemosensitive relapse of aggressive lym- median age at presentation is 65. In the UK,
phoma. The role of autologous stem cell trans- around 2000 new cases are diagnosed per annum.
plantation in relapse after rituximab-containing
front-line treatment has not been determined. Aetiology
The role of allogeneic transplantation in the The majority of acute leukaemias are sporadic,
management of patients with aggressive lym- although some genetic syndromes and other
phoma remains controversial. factors can predispose to acute leukaemia.
Recent advances • Genetic syndromes
The use of FDG-PET imaging in lymphomas is • Down’s syndrome
evolving. It is useful to define the extent of disease • Fanconi anaemia
308 as well as allow cessation of treatment when a • Neurofibromatosis
CANCERS OF THE HAEMATOPOIETIC SYSTEM 21
• Chemicals:
• benzene
• pesticides
• Previous chemotherapy (AML):
• alkylating agents
• topoisomerase II inhibitors
Pathology
AML and ALL are characterized by circulating
blasts cells which may be seen in the peripheral
blood. Bone marrow examination with immuno
phenotypic analysis is needed for establishing a
diagnosis. Both myeloblasts and lymphoblasts A
are characterized by a high nuclear : cytoplasmic
ratio and prominent nuclei. Azurophilic Auer
rods are pathognomonic of AML (Figure 21.6).
The French–American–British (FAB) classifica-
tion recognizes three subtypes of ALL: L1 (30%),
L2 (60%) and L3 (10%), whereas the current
WHO classification incorporates immunopheno-
typing and cytogenetics. By immunophenotyp-
ing, 75% ALL arise from B-cell progenitors and
25% from T-cells.
The FAB classification recognizes eight sub-
types of AML: M0 minimal myeloid differentia-
tion (3%), M1 poorly differentiated myeloblasts
(15–20%), M2 myeloblastic with differentiation
(25–30%), M3 promyelocytic (5–10%), M4 B
myelo-monoblastic (20%), M5 monoblastic (2–
Figure 21.6
9%), M6 erythroblastic (3–5%), and M7
AML M3 shows promyelocytes with multiple/stacked
megakaryoblastic (3–12%). The recent WHO Auer rods (A) and AML with a blast in the centre
classification incorporates cytogenetics and showing an Auer rod (B). Courtesy of Dr. Priyanka
classifies AML as: Mehta, Bristol Oncology Centre.
* Areas are liver, spleen or lymph nodes (either unilateral or bilateral) in inguinal, axillary and cervical region
313
II SITE-SPECIFIC CANCER MANAGEMENT
New agents
Oblimersen sodium, an antisense molecule to
Bcl-2 (Bcl-2 is an anti-apoptotic protein, is highly
expressed in CLL cells), in combination with Figure 21.7 Peripheral-blood smear showing
fludarabine and cyclophosphamide has shown to characteristic hairy-cell features. From Mey U, Strehl J
improve CR. Lenolidomide, an anti-angiogeneic et al. Advances in the treatment of hairy cell leukaemia,
agent, and flavopiridol, a cyclin dependent kinases Lancet Oncology 2003;4:86–94.
inhibitor, are shown to have activity in CLL.
two months followed by nucleoside analogue
Hairy cell leukaemia treatment.
Patients with a large spleen and with little
Hairy cell leukaemia (HCL) constitutes 2% of
or moderate bone marrow can be treated with
lymphoid leukaemias. The median age at presen-
splenectomy with delayed purine analogues until
tation is 55 years and males are commonly
progression.
affected. There is no known aetiological factor.
The majority (70%) of relapsed patients
One-quarter of patients are asymptomatic and
respond to retreatment with pentostatin or
diagnosis is made after an incidental finding
cladribine. In patients who fail to respond to
of splenomegaly or cytopenia. One-quarter of
nucleoside analogues, rituximab is useful.
patients present with abdominal symptoms due
to splenomegaly; one quarter of patients with
non-specific symptoms of fatigue, weight loss Myelodysplastic syndromes
and fever and the remainder present with fea- Myelodysplastic syndrome (MDS) is a heteroge-
tures of bleeding or recurrent infections. neous group of clonal stem cell disease character-
Peripheral smear shows cytopenia and the ized by dysplasia and ineffective haematopoiesis
presence of hairy cells, cells twice the size of a in one or major cell lineages. Although the
normal lymphocyte with cytoplasmic projections marrow is producing an excess of cells, these are
and an oval nucleus (Figure 21.7). Bone marrow immature and destroyed before reaching the
biopsy is important in definitive diagnosis, which circulation such that the peripheral blood appears
shows an interstitial or focal pattern of infiltra- hypocellular. The median age of occurrence is 70
tion. Immunophenotyping is necessary to distin- years with over 90% patients over the age of 50
guish HCL from other B-cell lymphomas. at the time of diagnosis.
Asymptomatic patients can be observed until There are a number of types including:
the development of cytopenia or systemic symp-
• Refractory anaemia
toms, when treatment is indicated. The mainstay
• Refractory anaemia with ring sideroblasts
of treatment is nucleoside analogues, Pentostatin
• Refractory anaemia with excess blasts
and cladribine, which give a complete remission
• Refractory anaemia with excess blasts in
of >80% with a 10-year overall survival of 95–
transformation
100%. Since both drugs can cause lymphopenia,
• Chronic myelomonocytic leukaemia
patients presenting with cytopenia are treated
either with G-CSF support during nucleoside In the majority of cases, MDS occurs as a de
analogue treatment or initial interferon alfa for novo disorder; recently secondary MDS/AML as
314
CANCERS OF THE HAEMATOPOIETIC SYSTEM 21
a result of chemotherapy/radiotherapy is increas-
ing. Radiotherapy and alkylating agent related
MDS occurs 5–6 years after treatment whereas
topoisomerase II inhibitor (e.g. etoposide and
teniposide) related MDS develops at a median
interval of 33–34 months after exposure.
Presentation depends on the associated cyto-
penia. Evaluation includes bone marrow biopsy
with iron stain and cytogenetic studies, erythro-
poietin levels and iron studies. The International
Prognostic Scoring System (IPSS) defines four
risk groups for overall survival and AML evolu-
tion, based on the percentage of marrow blasts,
specific cytogenetic abnormalities and number of
cytopenias. Treatment is complex which depends
on the subtype of MDS, IPSS, performance status
and co-morbidities which is not dealt with in this
text-book. In general the initial treatment is sup-
portive but can later involve chemotherapy or
even stem cell transplantation. Median survival
is related to IPSS score and ranges from a few
months to many years.
Figure 21.8
Solitary plasmacytoma MRI scan of the lumbo-sacral spine of a 69-year-old man
who presented with cauda equina syndrome due to L4
The incidence of solitary plasmacytoma is one- compression (arrow). Biopsy of L4 vertebra showed
tenth of myeloma. Solitary plasmacytomas can plasmacytoma and MRI showed another lesion in L1 and
changes in L2 (arrow heads), suggesting myeloma.
occur in bone (solitary bone plasmacytoma –
SBP) or in extramedullary sites (extramedullary
plasmacytoma – SEP). SBP commonly involves
the spine, ribs, femur, humerus, and skull and in all patients as up to one-third of patients may
SEP commonly occurs (80%) in the upper respi- have additional occult lesions that will be missed
ratory tract. In >50% cases solitary plasmacy- on skeletal survey (Figure 21.8). There may be a
toma is a precursor of myeloma. It is more monoclonal gammopathy, which has prognostic
common in men and the median age at diagnosis significance, in 24–72% of patients.
is 10 years younger than patients with multiple
myeloma. Treatment
Radical radiotherapy is the treatment for choice
Clinical features of SPB and SEP (Box 21.7). There is no role for
Pain is the most common clinical symptom of surgery in SPB in the absence of structural insta-
solitary bone plasmacytoma. Spinal plasmacy- bility or neurological compromise. Patients who
toma can present with spinal cord compression. need surgery receive postoperative radiotherapy.
Presentation of extramedullary plasmacytoma There is no data to recommend adjuvant chemo-
depends on the site of origin. therapy. Some consider chemotherapy for patients
at high risk of failure (e.g. tumour >5 cm).
Diagnosis Surgery is avoided in head and neck SEP.
The diagnostic approach is the same as that for Surgery may be considered for SEP at other
myeloma and solitary plasmacytoma is diag- sites. After a complete excision, radiotherapy
nosed when there is no evidence of myeloma. may not be necessary and patients with incom-
MRI of the spine and pelvis should be performed plete excision require radiotherapy. Adjuvant
315
II SITE-SPECIFIC CANCER MANAGEMENT
Figure 21.10
Symptomatic myeloma Candidates for ASCT
• Age 65 years or less Management of myeloma.
• Good PS
• No comorbidities
Potential candidate for ASCT
Practice in North America
Salvage therapy
12–32 months. Monitoring of asymptomatic Patients not eligible for ASCT are treated with
myeloma includes 3-monthly clinical assessment melphalan-based regimes (Figure 21.2 and Table
and measurement of paraprotein. 21.9). MPT is the preferred regimen for
Treatment of newly diagnosed multiple standard-risk patients who are not candidates
myeloma is rapidly evolving. The North Ameri- for transplantation. ASCT prolongs the median
can approach involves risk categorization of overall survival in myeloma by approximately 12
patients based on molecular cytogenetics and months.
upfront use of novel agents such as bortezomib
(a proteasome inhibitor) and lenalidomide (an Refractory and relapsing myeloma
analogue of thalidomide) whereas such an Almost all patients with myeloma eventually
approach is yet to be adopted in the UK (Figure relapse. If relapse occurs more than 6 months
21.10). Table 21.9 shows regimes in newly diag- after stopping therapy, the initial chemotherapy
nosed myeloma. Treatment response is assessed regimen should be re-instituted. Patients who
by the International Myeloma Working Group have cryopreserved stem cells early in the disease
definitions of response criteria. course can derive significant benefit from ASCT
In patients eligible for autologous stem cell as salvage therapy. In general, patients who have
transplantation (ASCT) prolonged melphalan indolent relapse can often be treated with single
based chemotherapy can interfere with adequate agents whereas those with more aggressive
stem cell mobilization and is therefore avoided. relapse often require a combination of agents.
318
CANCERS OF THE HAEMATOPOIETIC SYSTEM 21
meniningococci and Haemophilus influenza.
Table 21.9: Regimes in newly diagnosed Prompt treatment of infection is required.
myeloma
• Hyperviscosity syndrome needs plasma
Regimen Response rate
pheresis.
Regime for patients not eligible for ASCT
Melphalan–prednisolone–
thalidomide (MPT)
75%
Monoclonal gammopathy of
Bortezomib–melphalan- 70% unknown significance (MGUS)
prednisone (VMP) and smouldering myeloma
Cyclophosphamide– 72%
dexamethasone–thalidomide
MGUS is defined by a serum M-protein concen-
(CDT) tration of <3 g/dL, <10% plasma cells in the
bone marrow and absent myeloma-related organ
Regime for patients eligible for ASCT
or tissue impairment (ROTI). Around 1% per
Vincristine–Adriamycin– 52% year will transform to myeloma or another
dexamethasone (VAD)
plasma cell disorder. It does not require any
Thalidomide–dexamethasone 65% treatment but needs life-long monitoring (6–12
(Thal/Dex) monthly) with clinical assessment and quantifica-
Lenalidomide–low dose 70% tion of paraprotein.
dexamethasone (Rev/Dex) Smouldering myeloma is the stage between
Bortezomib–dexamethasone (Vel/ 80% MGUS and myeloma characterized by serum M
Dex) protein of ≥3 g/dL and/or ≥10% plasma cell in
Bortezomib–thalidomide– 90% the bone marrow and no ROTI. Transformation
dexamethasone (VTD) to myeloma or other plasma cell disorders occur
at a rate of 10% per year. Management is 3–4
monthly observation until evidence of progres-
sion to myeloma.
Supportive care Further reading
• Bone disease – all patients with symptomatic Evens AM, Hutchings M, Diehl V. Treatment of Hodgkin
myeloma receive regular bisphosphonate for lymphoma: the past, present, and future. Nat Clin Pract
at least one year from diagnosis. Oncol. 2008;5:543–556.
• Pain control is by analgesics and palliative Peggs KS, Anderlini P, Sureda A. Allogeneic transplantation
for Hodgkin lymphoma. Br J Haematol. 2008;143:
radiotherapy. Percutaneous vertebroplasty
468–480.
and kyphoplasty are useful for vertebral com- Diehl V, Fuchs M. Early, intermediate and advanced
pression fractures that do not respond to Hodgkin’s lymphoma: modern treatment strategies. Ann
standard measures within 6 weeks. Oncol. 2007;18 Suppl 9:ix71–79.
• Hypercalcaemia is treated with Lenz G, Staudt LM. Aggressive lymphomas. N Engl J Med.
2010;362:1417–1429.
bisphosphonates.
Zucca E. Extranodal lymphoma: a reappraisal. Ann Oncol.
• Renal impairment: managed with adequate 2008;19 Suppl 4:iv77–80.
hydration, avoidance of nephrotoxic drugs Estey E, Döhner H. Acute myeloid leukaemia. Lancet.
and prompt treatment of infection and 2006;368:1894–1907.
hypercalcaemia. Hehlmann R, Hochhaus A, Baccarani M. Chronic myeloid
leukaemia. Lancet. 2007;370:342–350.
• Anaemia – transfusions, erythropoietin or
Shanafelt TD, Kay NE. Combination therapies for previously
darbepoietin are useful. untreated CLL. Lancet. 2007;21;370:197–198.
• Infections – all symptomatic patients need Rajkumar SV. Multiple myeloma. Curr Probl Cancer.
vaccination against influenza, pneumococci, 2009;33:7–64.
319
Paediatric, teenage and
young adult cancers
HM Hatcher
22
Introduction Aetiology
Cancers in this group of patients present specific The majority of older adult cancers are related
challenges in diagnosis, treatment, recruitment to to specific risk factors such as smoking, whereas
clinical trials and survival. Childhood cancers the majority of paediatric malignancies are
have seen a significant improvement in survival thought to be developmental in origin. A number
in the last 30 years from less than 30% to greater of syndromes are associated with childhood
than 70% long-term survival. Unfortunately the cancer (e.g. Down’s syndrome associated with
same is not the case in the teenage and young leukaemia) but the majority arise without an
adult cancers (TYA) with very little improvement underlying genetic predisposition (e.g. Li–
seen over the same time period. Fraumeni and sarcomas). TYA malignancies fall
In addition they have particular social, educa- between the two age extremes and may represent
tional, developmental and psychological needs a late developmental malignancy or an early adult
which, if not addressed, have long-term conse- malignancy due to other factors, e.g. familial
quences for the patient and their family. Those adenomatous polyposis. In most cases the
who survive cancer at a young age may also be aetiology is not known.
left with significant medical late effects which
need to be recognized and managed (p. 58).
Types of cancer
Incidence/epidemiology The types of cancer vary not only between chil-
dren and adults but as the age increases in certain
The incidence of malignancy in the UK is: age group bandings (Table 22.1).
• 12.4 per 100,000 up to 15 years. In children leukaemias represent the most
• 14.4 per 100,000 in those 15–19. common malignancy with acute lymphoblastic
• 22.6 per 100,000 in those 20–24. leukaemia (ALL) being most frequent. Brain
tumours also make up a significant group, but
Those aged 14–24 account for 0.5% of all
the incidence falls as a proportion of cases from
cancer registrations in the UK. In a study from
childhood towards early adulthood. In the 15
the north-west of England the incidence was 174
to 19-year-olds, the most frequent malignancies
cases per million with a male : female ratio of
are lymphomas, leukaemias and carcinomas
1.22 : 1. In the same study the incidence was
(especially thyroid and nasopharyngeal). For the
found to be increasing, particularly for bone
20 to 24-year-olds, lymphomas remain the most
tumours, testicular tumours, thyroid cancer and
common malignancy but with carcinomas (espe-
malignant melanoma.
cially cervix, thyroid and breast) and germ cell
Cancer is the most frequent natural cause of
tumours as next common. Osteosarcomas and
death in this age group, second only to accidents.
germ cell tumours peak in late teenage and
• Male : female ratio is 1.2 : 1. early adulthood.
320 © 2011, Elsevier Ltd
DOI: 10.1016/B978-0-7234-3458-0.00027-0
PAEDIATRIC, TEENAGE AND YOUNG ADULT CANCERS 22
The clinical features in children, teenagers and • Obvious lump or swelling around the head, neck,
abdomen or elsewhere.
young adults are similar to those seen in other
• Increasing and persistent pain in bone or joints,
age groups with the same malignancies, but are often leading to less use of the limb or limping.
often not recognized as such due to the rarity of • Pallor, especially if associated with unexplained
malignancy in these age groups. In addition for bruising or bleeding.
children who are unable to explain symptoms • Worsening headaches, especially if associated with
clearly, the disease has usually progressed so that vomiting in the morning.
symptoms are obvious to their family. Delays in
diagnosis are very common in the adolescent and
young adult and can contribute to reduced
survival at all ages. Delays in diagnosis can occur in children if
In children warning signs may be due to local- symptoms are occult, especially in very young
ized disease, or the manifestations of dissemi- children but are more common in the TYA group.
nated disease are listed in Box 22.1.
In the older child and young adult, the Specific childhood cancers
same warning features apply but also include
additional signs specific to the different Some cancers are specific to children and young
malignancies. adults, or their treatment is different to that in
adults. These tumours will be highlighted below.
Diagnosis and delays For other malignancies, covered by other chap-
ters, only specific changes in management for
Diagnosis of malignancy currently follows the children and young adults will be made.
pathways for either paediatric or adult cancers.
Paediatric cancers are diagnosed and treated in Leukaemias
specialist principal treatment centres with some In childhood acute lymphoblastic leukaemia
care shared with local hospitals. For TYA cancers, (ALL) is by far the most frequently occurring
currently they may be seen in a paediatric or leukaemia representing over 75% cases. (p. 308).
adult setting and treated according to those pro- Presentation is usually with symptoms and
tocols. In the UK TYA cancer will soon be treated signs of bone marrow infiltration and these can
in specialized centres. This may mean referral to appear to occur quite rapidly, although are often
a specialist TYA centre or to an adult cancer preceded by several weeks of general malaise or
specialist in a particular site of the body in a recurrent infections and fevers. Usual features at
designated regional hospital with appropriate the time of diagnosis are pallor, abnormal bruis-
TYA facilities (Figure 22.1). ing or bleeding, bone pain (which may manifest
321
II SITE-SPECIFIC CANCER MANAGEMENT
Figure 22.1
Suspected cancer
Referral for treatment of childhood
and TYA malignancy.
Refer to specialist
paediatric oncology
centre for diagnosis
Rare Common
as limping in young children) and on clinical Treatment of childhood and young adult ALL
examination lymphadenopathy and hepato is performed according to risk stratification as in
megaly in some children. Box 22.2. In general treatment involves several
Diagnosis is suspected on the differential blood phases which is similar to that of adult ALL
count but bone marrow examination is essential (p. 308).
to confirm the diagnosis (p. 308), to perform Relapsed ALL may still be curable with high-
immunocytochemical stains and cytogenetics dose chemotherapy and stem cell transplanta-
which are of diagnostic and prognostic signifi- tion, but the chances of long-term cure are
cance (Box 22.2). much lower with the high-risk subgroups and if
322
PAEDIATRIC, TEENAGE AND YOUNG ADULT CANCERS 22
relapse is within a year of completing initial Overall more than 80% are cured of their disease.
treatment. Treatment for relapse is rarely curative.
In addition certain subgroups such as B-cell
ALL are often treated on different regimens Neuroblastoma
similar to those used for a Burkitt’s lymphoma Neuroblastoma is a neuroendocrine tumour
(p. 305, Burkitt’s lymphoma). Both these malig which originates from any neural crest element
nancies are associated with a t(8;14) translocation of the sympathetic nervous system. It predomi-
so may be variations of the same malignancy. nantly occurs in those under 3 years of age. The
highest incidence is in the first 12 months of life
Brain tumours with less than 10% of cases occurring in those
Brain tumours are the second most common over the age of 5 years.
malignancy in children and remain a significant Clinical presentation is varied depending on the
cause of cancer into young adulthood. In location and extent of the tumour. Fatigue, fever
children the majority of tumours occur infra and anorexia are the most common initial pre-
tentorially but this proportion changes with senting features with more localizing features
increasing age. dependent on tumour site. Adrenal primaries are
Presenting features include headaches, often often extensive at presentation and may involve
worse in the morning and associated with the IVC. Paravertebral tumours may lead to spinal
morning vomiting, papilloedema (sometimes cord compression. At least 50% of cases are meta-
detected by an optician), ataxia, personality static at presentation. Elevated catecholamines
change, nerve palsies and nystagmus. are found in over 90%. Staging includes a CT,
Common subtypes of brain tumour include bone scintigram, bone marrow sampling and
astrocytoma, medulloblastoma, glioma, cranio radiolabelled MIBG (meta-iodobenzylguanidine)
pharyngioma and ependymoma. In children, scan. MIBG is taken up by 95% of neuro
particularly in the very young, the emphasis of blastomas and acts as a tool for staging and
treatment is to delay, avoid or minimize the need monitoring disease response.
for radiotherapy to avoid long-term side effects. Poor prognostic features are advanced stage,
These are covered in more detail on p. 264. age over 1 year, and over-expression of N-myc
oncogene.
Wilms’ tumour Treatment may be with surgery alone in early
Wilms’ tumour or nephroblastoma occurs most stage disease, but the majority require combina-
frequently in children under 5 years of age and tion chemotherapy. Trials are examining the role
very rarely in older children or young adults. of intensification of treatment in poor prognosis
Most tumours are unilateral, but are bilateral in disease. The risk of relapse is high and the long-
5% of cases and in a further 5–10% will have term survival for those with metastatic disease at
more than one tumour in the same kidney. Risk presentation is less than 30%.
factors include certain genetic syndromes such as
Beckwith–Weidemann syndrome. They are often Lymphomas
asymptomatic until they present as an abdominal Lymphomas occur in childhood and Hodgkin’s
mass, pain and/or anorexia. disease has a peak in incidence in young adult-
Histologically they are divided into a favoura- hood (p. 298). In general these tumours are
ble group with well-defined elements and the ana- treated similar to their adult counterparts
plastic (or unfavourable group) with poorly except that radiotherapy is avoided, except in
defined cellular morphology. Less than 25% have refractory disease, to reduce the risk of its effect
a mutation within the WT1 (Wilms’ tumour) gene. on growth and the risk of second malignancy.
Staging is undertaken with imaging and Chemotherapy regimens are similar to adults but
treatment is according to stage (Figure 22.2). Ana- trials are ongoing to assess the role of rituximab
plastic tumours are treated with more aggressive in children. The most common subtypes of NHL
chemotherapy except in stage I where their prog- in children are Burkitt-like lymphomas (p. 305)
nosis is similar to favourable histology tumours. and diffuse large B-cell lymphomas (Table 22.2).
323
II SITE-SPECIFIC CANCER MANAGEMENT
Staging
Ultrasound-blood vessel
involvement CT, bone scan
(surgery)
work up for treatment:
bloods, MUGA/ECHO, GFR
Limited to the Tumour extends Unresectable primary Lung, liver, Bilateral tumours
kidney beyond the kidney or LN metastases or bone, brain Both sides should
Completely excised but excised extensive or distant LN be staged
Intact capsule May be tumour spillage tumour spillage metastases
4 year survival 4 year survival 4 year survival 4 year survival 4 year survival
98% favourable 96% favourable 95% favourable 90% favourable 76–94%
85% anaplastic 70% anaplastic 56% anaplastic 17% anaplastic
Figure 22.2
Staging and management of Wilms’ tumour.
324
PAEDIATRIC, TEENAGE AND YOUNG ADULT CANCERS 22
Anaplastic large cell tumours are rare as are but due to the aggressive nature of these tumours,
other adult subtypes such as cutaneous T-cell combination chemotherapy with e.g. IVADo
lymphoma and MALT lymphoma (p. 306). Prog- (ifosfamide, vincristine, actinomycin and doxo-
nostic factors are similar to adult tumours. rubicin), is given. Radiotherapy is often given if
margins are positive or there has been tumour
Sarcomas regrowth after initial surgery. An Italian pilot
Both bone and soft tissue sarcomas occur in study suggested that maintenance therapy with
children and young adults. These are covered in cyclophosphamide and vinorelbine may prolong
detail in Chapter 15. Some sarcomas, however, progression free survival. In advanced disease
peak in this age group. Osteosarcomas peak in combination chemotherapy is used but the
early adolescence with Ewing’s sarcomas having outlook is poor. Poor prognostic factors include
their peak incidence in older adolescence. increasing age, particularly over age 16, advanced
Rhabdomyosarcoma (RMS), a tumour of stri- stage and alveolar subtype.
ated muscle, is the only soft tissue sarcoma to
peak in children and adolescents. It is divided Germ cell tumours
into two main histological subgroups, embryonal Germ cell tumours of the ovary and testes peak
and alveolar. Alveolar rhabdomyosarcoma in young adulthood and are described in more
(ARMS) is characterized by one of two specific detail in Chapters 12 and 13. Similar treatment
translocations, either t(2;13)(q35;q14) resulting modalities are used in children and young adults,
in fusion of PAX3-FOXO1 in 60% ARMS or but there has been a move to reduce long-term
t(1;13)(q36;q14) resulting in PAX7-FOXO1 in side effects from chemotherapy and to try to
20% ARMS. Embryonal RMS is not associated maintain fertility in ovarian germ cell tumours.
with a specific translocation, but loss in chromo- Trials are underway to examine the role of car-
some 11. Common tumour sites include the head boplatin rather than cisplatin and to reduce the
and neck, paratesticular and limb (Figure 22.3). dose of bleomycin in adolescent germ cell tumours
It frequently presents with advanced disease. If to reduce late effects. Fertility sparing surgery is
localized it can present with a mass or proptosis preferred if possible for ovarian germ cell tumours
if retro-orbital. It can give rise to nodal metas- and many women retain their fertility with this
tases and regional lymph nodes should be exam- type of surgery and BEP chemotherapy (p. 220).
ined. Staging involves local MRI, CT of chest
(and regional), bone scan, and bone marrow Langerhans cell histiocytosis (LCH)
biopsy. Localized disease is treated with surgery,
LCH is a proliferative disorder of histiocytes.
Although not strictly a malignancy it may exhibit
aggressive behaviour and require combination
chemotherapy. Multiple bony lesions can occur
at any site, but can occur within the skull leading
to hypothalamic infiltration and diabetes insip-
idus. Systemic LCH tends to present in infancy
as a widespread rash with bone marrow and soft
tissue involvement. Organ dysfunction is a better
indication of poor prognosis than the number of
involved sites. Treatment of advanced disease is
with chemotherapy.
Hepatoblastomas
Hepatoblastomas are rare but are most common
Figure 22.3
MRI of sagittal section through the foot showing a mass
malignancy of liver in children. There is an asso-
deep to the plantar fascia. Histology showed an ciation with FAP. Hepatocellular carcinoma can
embryonal rhabdomyosarcoma. also occur rarely but in older age groups. Clinical
325
II SITE-SPECIFIC CANCER MANAGEMENT
presentation is usually with a mass or abdominal an often protracted treatment period. The fact
distension. Jaundice is rare. Alpha-foetoprotein that many children are now cured of their disease
(AFP) is elevated in most cases. Unlike adult liver but with long-term effects from their treatment
tumours, most hepatoblastomas respond to also has a psychological impact on them and
combination chemotherapy (cisplatin and doxo- their family. Adolescents and young adults are at
rubicin) with good long-term survival. a vulnerable stage of development and treatment
for cancer has been shown to have a detrimental
Retinoblastoma long-term psychological impact in many patients.
Retinoblastomas are very rare childhood Appropriate support should be offered to patients
tumours. All bilateral tumours and 20% of uni- and their families to try to prevent some of these
lateral tumours are thought to be hereditary. The conditions occurring later on.
retinoblastoma tumour suppressor gene is on Risk-taking behaviour is also at its greatest in
chromosome 13 with the pattern of inheritance the TYA age group and can affect drug compli-
being autosomal dominant with incomplete pen- ance as well as coping mechanisms.
etrance (Chapter 5, p. 55). Most cases present
before age 3. In children not known to be in a Body image and fertility
retinoblastoma family presentation is with a
squint or white papillary reflex. Those related to Sexual identity and future fertility are important
retinoblastoma families should be part of a considerations for the adolescent or young adult.
screening programme. Surgery may be necessary Loss of hair or a limb will significantly affect
for some but most cases are treated with radio- body image at an age when this is especially
therapy. Long-term survival is over 95%, but vulnerable. The type of cancer or its treatment
second malignancy is common (osteosarcomas in may also affect future fertility, but this is not
radiotherapy field) and in those with hereditary universal so specific knowledge is required for
retinoblastoma other malignancies, especially each patient (p. 59, late effects).
sarcomas can occur elsewhere in the body in
young adulthood up to middle age. Education
The majority of children and young adult patients
Clinical trials will be in full-time education at the time of diag-
Traditionally children have a high recruitment to nosis. If treatment is lengthy or affects the ability
clinical trials (>80%) and there are often avail- to study in the longer term, this can also result
able trials in the most frequent childhood cancers. in difficulties reintegrating into school or obtain-
However, trial recruitment is lowest in the ado- ing a job. Continuing education should be sup-
lescent and young adult population when com- ported throughout treatment.
pared with either children or older adults. This
is partially due to the rarity of cancers concerned, Late effects
but also to the fact that these patients are treated
in a wide range of settings. Many fall between Late effects are an important factor in the treat-
different groups of physicians who may not be ment of children and young adults (p. 58, late
able to provide available trials. It is hoped this effects). Late effects are dependent upon the orig-
will be changed with the evolving changes in the inal cancer, its treatment, the family genetics and
organization of TYA cancer management. the developmental stage of the individual when
treated for cancer. Every organ system can be
affected, but the most significant include the risk
Psychological support of second malignancy, neurocognitive impair-
Children and their families frequently need ment, growth problems, and cardiac and endo-
support to help them manage the consequences crine abnormalities. Many centres run late effects
of a diagnosis of cancer at such a young age, and clinics to monitor for these and other sequelae of
the consequences it will have on the family over cancer treatment.
326
PAEDIATRIC, TEENAGE AND YOUNG ADULT CANCERS 22
327
Oncologic emergencies
J Wrigley and TV Ajithkumar
23
Metastatic spinal tantly, around 17% of patients have two or more
levels of compression at presentation.
cord compression Spinal cord metastases can be of three types
Introduction depending on the location of tumour deposit:
Metastatic spinal cord compression (MSCC) • Extradural – occurs in more than 90%,
occurs in 3–5% of patients with cancer. Prostate, usually due to direct extension of vertebral
breast and lung cancer each account for 15–20% metastases. 75% of these are caused by soft
of cases; lymphoma, myeloma and renal cell tissue extension and 25% by bone collapse
cancer account for 10% and the remainder is due and compression by bony fragments.
to colorectal cancers, sarcomas and cancers of • Intradural extramedullary – occurs in 5% of
unknown primary. Although more frequent in cases and is due to deposit via CSF spaces.
patients with known malignancy, MSCC is the • Intramedullary (0.5–3.5%) – are commonly
presenting feature in up to 20% of patients with associated with brain metastases and lepto
no prior diagnosis of cancer, particularly lung meningeal disease and can occur in lung,
cancer. breast and renal cell cancers, lymphoma and
myeloma.
Aetiology
The mechanisms by which MSCC may develop Clinical features
include: Back pain is the most frequent (95%) first
symptom, which commonly precedes the diagno-
1. Direct tumour extension into the epidural
sis of MSCC by up to 3 months in patients with
space from a vertebral or paravertebral mass.
known cancer and up to 5 months in those
2. Haematogenous spread to the vertebral spine
without a known malignancy. The pain may be
causing collapse and compression.
localized or neurogenic and is typically worse
3. Direct deposition of tumour cells within the
when the patient is supine.
cord.
Limb weakness is the second most common
Early stage compression of the cord results in symptom. Up to 85% of patients will have
oedema, venous congestion and demyelination, abnormal power in the limbs at presentation,
and neurological recovery may still be possible which depends on the neurologic level of
during this phase with prompt decompression. compression.
Continued compression causes secondary vascu- Sensory deficit may manifest itself as paraes-
lar injury and irreversible infarction. The most thesia or sensory loss and is present in up to 65%
common location of compression is thoracic of patients with MSCC. Up to 50% of patients
(50–70%) followed by lumbo-sacral (20–30%) will have a dermatomal sensory level, although
and cervical spine (10–20%). Up to 85% patients this may vary by several dermatomes above or
have multiple vertebral metastases and impor- below the true level of compression.
328 © 2011, Elsevier Ltd
DOI: 10.1016/B978-0-7234-3458-0.00028-2
ONCOLOGIC EMERGENCIES 23
Autonomic dysfunction is a late complication when MRI is contraindicated. A sagittal screen-
affecting up to 50% of patients with MSCC. ing of the whole spine with T1 and short T1
It includes impotence, bladder and bowel inversion recovery (STIR) images should be done
dysfunction, with constipation occurring most to ensure that multiple levels of compression are
frequently. not missed and also to identify asymptomatic
There are two clinical variants of MSCC, metastases. T2 weighted images help to detect
which require early recognition: the degree of compression by a soft tissue com-
ponent and intramedullary lesions (Figure 23.1).
• Isolated ataxia: thoracic SCC may present
In patients who have not been previously
with isolated ataxia due to spinocerebellar
diagnosed with cancer, a histological diagnosis
pathway dysfunction.
is required before definitive treatment can be
• Cauda equina syndrome: epidural extension of
planned. Patients should be investigated like
metastases below the level of the spinal cord
any other patient with cancer of unknown
(L1/L2) can present with early sphincter dys-
primary and an appropriate area most amenable
function, saddle anaesthesia (buttocks and
to biopsy should be established. However, treat-
perineal region), flaccid paraparesis of hip
ment should not be delayed during these investi-
extension and abduction, knee flexion and
gations and prompt neurosurgical involvement
movement of feet and toes, and absent ankle
should be established if biopsy is not possible.
reflexes.
Investigations Management
MRI of the whole spine is the investigation of The aim of treatment is to preserve or improve
choice for suspected MSCC. CT is used only neurological function and achieve pain control.
A B
Figure 23.1
Imaging in cord compression. MRI scan shows
spinal cord compression due to a collapsed
C vertebra (A,B) and an intramedullary
metastasis from lung cancer (C).
329
II SITE-SPECIFIC CANCER MANAGEMENT
70–100% patients who are ambulant at the Box 23.1: Patients with MSCC according to
beginning of treatment remain ambulant with outcome after surgical intervention
prompt treatment, whereas only 30–50% patients Good surgical candidates Poor surgical candidates
who are non-ambulant regain the ability to walk ‘Good prognosis tumours’ ‘Poor prognosis tumours’
and only 5–10% of paraplegic patients become e.g. breast cancer, e.g. lung cancer,
ambulant. Hence it is important to have defini- testicular cancer etc. melanoma
Single level of compression Multiple levels affected
tive treatment within 24 hours of presentation with solitary or few or multiple spinal
with suspected cord compression. Patients with vertebral metastases metastases
MSCC secondary to a vascular event will not Absence of visceral Presence of visceral
metastases metastases
respond to treatment. Good neurological function Poor neurological
function
Initial measures No previous radiotherapy Recurrence following
radiotherapy
All patients with suspected MSCC, except those Minimal co-morbidity Medically unfit for
without known cancer and those who are likely surgery
to have lymphoma, are started on oral dexame- Unknown primary or no Prognosis <3 months
histopathological
thasone 16 mg/day after assessment. This dose of diagnosis
steroid is continued until definitive treatment,
after which the steroids are weaned down as
quickly as neurological symptoms will allow.
Optimal pain control and cervical collar for Box 23.2: Indications for surgical treatment*
patients with suspected cervical spine involve- • Relapse after radiotherapy
ment are advised. • Progression while on radiotherapy
• Radiation resistant tumours
Specific measures
• Unknown primary tumour
Definite treatment of MSCC depends on the • Unstable spine or pathological fractures
histologic type and associated spinal stability. In
patients with no prior history of cancer, surgical *With a single site of cord compression and no total paraplegia for
longer than 48 h.
decompression with histologic confirmation is
appropriate. If surgical decompression is not
possible, a CT guided biopsy is needed.
Surgery may involve decompression, stabiliza- Box 23.3: Radiotherapy in spinal cord
tion and or resection and reconstruction of the compression
spinal canal. The patient’s overall prognosis and • Position – supine or prone
performance status should be taken into consid- • Treatment volume – one vertebra above and
eration and patients who have had no distal below MRI proven compression
neurological function for >24 h should not be • Ensure that all paravertebral disease is enclosed in
the volume if feasible
considered for surgery (Boxes 23.1 and 23.2).
• Direct posterior beam 6 MV (parallel lateral in
Radiotherapy is the treatment most frequently cervical region if feasible)
used and is most effective for patients with radio- • Prescription point – anterior spinal canal (either
sensitive tumours who are ambulatory at the measured from MRI or arbitrary, see Figure 23.2)
beginning of treatment. For those without • Dose – 30 Gy in 10 fractions or 20 Gy in five
mechanical pain or structural instability, radio- fractions
therapy may significantly improve pain control
and neurological function. The most commonly
used regimes are 20 Gy in five fractions (more Patients with established paraplegia are treated
appropriate for patients with expected short sur- with an 8 Gy single fraction for pain control.
vival) or 30 Gy in 10 fractions (Box 23.3). Some The issue of whether surgery or radiotherapy
patients may deteriorate during radiotherapy or a combination of both gives best functional
when the steroid dose may be increased or they outcome is yet to be resolved. A randomized
may be considered for surgery if appropriate. study compared radiotherapy (30 Gy in 10 frac-
330
ONCOLOGIC EMERGENCIES 23
Figure 23.2
Radiotherapy in spinal cord
compression – anatomical
landmarks and arbitrary
radiotherapy dose prescription
points.
Cervical
Superior thyroid
notch C4 C6 – 7 cm
Sternal notch T2 T2 – 7 cm
Sternal angle T4
T5 – 6 cm
Thoracic
T8 – 5 cm
Xiphisternum T9
T11 – 6 cm
Sacral
tions) started within 24 hours of onset of MSCC unpredictable. Hence MSCC in chemosensitive
with surgery within 24 hours followed by radio- tumours is treated with a combination of radio-
therapy within 2 weeks of surgery. Results therapy and chemotherapy.
showed that initial surgery followed by radio-
therapy offers a longer period with ability to Supportive measures
walk compared with those treated with radio- Supportive care requires a multidisciplinary team
therapy alone (median, 126 days vs. 35 days, approach. Analgesia needs to be optimized and
p = 0.006). This study showed that surgery venous thromboembolism prophylaxis pre-
permits most patients to remain ambulatory and scribed. Urinary or supra-pubic catheters should
continent for the remainder of their lives, while be considered and faecal continence should be
patients treated with radiation alone spend controlled with the use of stool softeners and
approximately two-thirds of their remaining time suppositories every 2–3 days. In patients with
unable to walk and incontinent. However, results bone metastases, there is no evidence that
of this study may not be extended to all patients bisphosphonates can reduce the risk of cord
as the study was limited to patients with less compression.
radiosensitive tumours and with different tumour
types and presentations. Prognosis
Chemotherapy alone is not an option for The median survival following a diagnosis of
treatment even in chemosensitive tumours as MSCC is 3–6 months, with 17% of patients
the response to treatment is often slow and alive at 1 year and 10% alive at 18 months.
331
II SITE-SPECIFIC CANCER MANAGEMENT
Pre-treatment motor function is an important neurological deficits. Seizures and bleeding into
predictor of the ambulatory outcome. Patients the metastases can result in an acute medical
who develop motor dysfunction more slowly also emergency. Malignant melanoma, renal cell car-
may have a better outcome. cinoma, thyroid cancer and choriocarcinoma are
commonly associated with bleeding into the
Recurrence of spinal cord compression metastases. Initial treatment is dexamethasone
7–14% of patients can present with recurrence 16 mg daily. Patients who present with seizures
of spinal cord compression after initial decom- require anticonvulsants. Role of prophylactic
pression. If the second cord compression is in a anticonvulsants is not known; it may be consid-
different area, treatment is the same as that ered in patients with high risk features of seizure
of the original compression. However, if patients such as metastasis to the motor cortex, bleeding
present with spinal cord compression at the metastasis and leptomeningeal metastasis. Further
initial radiation portal, surgical decompression is management of brain metastasis is given on p. 278.
the initial option, particularly if the recurrence is
within three months of initial treatment. If they Encephalopathy
are not a candidate for surgery, re-irradiation
Introduction
may be considered. It is important to make sure
that the total dose to spinal code is kept below Encephalopathy is an important complication of
100 Gy2 (biologically equivalent dose of 100 Gy cancer, which presents with an acute confusional
when delivered as 2 Gy per fraction). Many clini- state or delirium subsequent to abnormal brain
cians use a dose of 20 Gy in 8–10 fractions. function resulting from interference with brain
metabolism. A number of aetiological factors
Paediatric spinal cord compression can contribute to the development of encepha-
Paediatric MSCC differs from adult MSCC in that lopathy in cancer patients. Generally, these are
it is often caused by chemosensitive histological due to infection, metabolic abnormalities (such
types not seen in adults such as neuroblastoma, as hyponatraemia, hypercalcaemia) and drugs. A
Wilms’ tumour (p. 323). The usual pathogenesis number of anti-neoplastic agents such as ifosfa-
is the direct invasion of tumour through neural mide, methotrexate, cisplatin, vincristine, cyto-
foramina. The most common histologic type is sine arabinoside etc. can cause encephalopathy.
neuroblastoma, which responds to chemotherapy. Clinical features
Decompressive surgery is offered when patients The usual presentation is cognitive and behav-
present with rapid progression or progress during ioural changes. Patients can present with insom-
chemotherapy. Radiotherapy is only offered when nia followed by acute confusion and delirium.
there is no response after chemotherapy and/or Occasionally focal signs such as ataxia may
surgery and for those who require palliation after predominate.
failure of multiple systemic regimens. Clinical examination shows an altered level
of consciousness, abnormalities of respiration,
Intramedullary spinal cord metastasis
small reactive pupils and spontaneously roving
This is rare (1% incidence) and lung cancer is eye movements. Grading of encephalopathy is as
the most common primary. Sensory deficit (79%), follows:
sphincter dysfunction (60%) and weakness
(91%) are common manifestations. Up to 40% • G1 – mild transient drowsiness or nightmares.
patients will have brain metastases. Treatment is This may not be noticed by the patient at the
with corticosteroids and radiotherapy. time of treatment, but nightmares may be
reported at the next clinic visit.
• G2 – drowsy <50% time.
Raised intracranial pressure • G3 – drowsy >50% time, severe disorientation,
Approximately 25% of patients with cancer tremor, psychosis. Patients will often present
develop brain metastases and can present with with 1 or 2 of these features but not all.
features of raised intracranial pressure and focal • G4 – coma or seizures.
332
ONCOLOGIC EMERGENCIES 23
with a neutrophil count ≤1.0 × 109/l have (especially in those aged <45 years) and there-
bacteraemia. fore, prompt assessment is essential. Due to lack
Susceptibility to infection increases as the of neutrophils, most infections present with atyp-
neutrophil count drops below 1.0 × 109/l. The ical manifestations.
frequency and severity of infection are inversely It is important to enquire and look for signs
proportional to the absolute neutrophil count, of infection at the following sites:
with the duration of neutropenia also contribut-
• Head and neck – look at the teeth, gums and
ing to overall risk. The timing of the neutrophil
pharynx. Ask about ENT problems, espe-
nadir depends on the type of chemotherapy and
cially involving the sinuses and ears.
generally, it occurs 5–10 days after the last dose.
• Gastrointestinal system – ask about mucosi-
Usually the neutrophil count recovers 5 days
tis, diarrhoea and constipation. The perineal/
after the nadir.
peri-anal area should be inspected and pal-
In the majority of cases, bacterial pathogens
pated gently; but do not perform a rectal
are responsible for febrile neutropenic episodes
examination.
with fungal (Figure 23.3), viral and protozoal
• Respiratory system – ask about cough, short-
infections occurring more commonly as second-
ness of breath and sputum production.
ary events. Currently, Gram-positive bacteria
• Genito-urinary system – ask about symptoms
account for 60–70% of microbiologically
suggestive of infection such as vaginal
detected infections, which may in part be due to
discharge, abdominal pain, urinary frequency
the prevalent use of quinolones as prophylactic
and dysuria.
antibiotics. Other possible causes of this change
• Central nervous system – ask about headache.
in trend include widespread use of intravenous
Examine for altered level of consciousness,
catheters, along with more profound and pro-
cranial nerve lesions and meningism.
longed neutropenia due to intensive and recur-
• Look at vascular access sites and establish
rent treatment regimes.
whether symptoms are related to flushing of
Clinical presentation and assessment the device.
Fever, rigors, hypotension or generalized malaise Antibiotic treatment should not be delayed
may be the only presenting features of infection whilst waiting for results; however, urgent inves-
in the neutropenic patient, and even they may tigations should include:
be masked by concurrent use of NSAIDs or
• FBC and differential, U+E, CRP, LFTs, Ca2+,
steroids. Clinical deterioration can be rapid
coagulation screen and group and save.
• Blood cultures should be taken both peripher-
ally and through any central venous line.
• Stool, urine and throat swabs should be sent
for microscopy, bacterial culture and sensitiv-
ity (viral culture also for throat swab).
• Skin, wound or genital swabs should be
collected when appropriate.
• Sputum sample if possible.
• Chest X-ray.
Investigations
Box 23.5: Indications for G-CSF
A plain chest radiograph will often show a right
paratracheal mass or widened mediastinum. A
Prophylactic Germ cell tumour
1. Regimes associated VeIP CT scan will demonstrate SVC compromise
with >20% risk of FN
2. Regimes with 10–20%
and distinguishes between external compression
Breast cancer risk of FN and with and thrombus (Figure 23.5). In patients without
AC followed by factors that may increase a histologic diagnosis, a biopsy is required prior
decetaxel the risk of FN to ≥20%.
The risk factors are:
to deciding treatment.
Paclitaxel followed by AC
TAC • Age ≥65 years
• Advanced disease Management
Lung cancer
Cisplatin-etoposide • Prior FN The management of SVCO depends on the
ACE • No antibiotics underlying aetiology and severity of symptoms.
prophylaxis
ICE In patients with no prior diagnosis of cancer, a
• Multiple
Ovarian cancer co-morbidities histologic diagnosis is essential.
Paclitaxel • Bone marrow General measures include oxygen, dexametha-
Docetaxel involvement sone 16 mg daily, and diuretics. Endovascular
NHL Therapeutic use of G-CSF stent insertion (Figure 23.5) is the gold-standard
DHAP during an episode of treatment which relieves obstruction in 95% of
ESHAP febrile neutropenia cases, usually within 72 h (Box 23.6). Compli
Patients with ongoing
CHOP-21 neutropenia who do cations of stent placement are in the region
Urothelial not respond to of 3–7% and include: transient chest pain,
Carboplatin/paclitaxel antibiotics infection, misplaced stent, stent migration and
BOP-VIP-B Those with life- pulmonary emboli. SVCO recurs in approxi-
threatening infections
mately 11% of patients (due to stent occlusion
• Shock
secondary to either thrombus or tumour in-
• Severe sepsis
growth). However, secondary patency rates are
• Multi-organ
dysfunction good and long-term patency is achieved in 92%
of patients.
337
II SITE-SPECIFIC CANCER MANAGEMENT
Life-threatening haemoptysis
Approximately 10% of haemoptysis due to
cancers tends to be massive (>500 ml in 24
hours) and <5% of haemoptysis is life-threaten-
ing. The reported in-hospital mortality of bleed-
ing at a rate of >1 litre in 24 hours is 80%. It is
difficult to predict life-threatening haemoptysis
and most patients die suddenly at home.
The immediate management of patients with
B
life-threatening haemoptysis is to maintain a
Figure 23.5 secure airway, adequate ventilation and circula-
Superior vena caval obstruction. CT scan (A) shows tion. Attempts should be made to localize the site
significant compression of SVC (arrow) and SVC stent in of bleeding with bronchoscopy (which can be
situ (B). therapeutic) and/or imaging. Once bleeding is
localized the patient is positioned with the bleed-
ing lung in the dependent position.
Radiotherapy was traditionally used first line Specific measures to control bleeding include
for the treatment of SVCO prior to endovascular bronchoscopy with tamponade using a balloon
stenting. In radiosensitive tumours, radiotherapy catheter, laser photocoagulation or iced-saline
results in symptomatic response rates of up to lavage. Bronchial artery embolization is an
78% at 2 weeks. However, complete resolution option in specialist units.
of obstruction is seen in only 31% on serial veno-
grams with a partial resolution in 23%. Central airway obstruction
Radiotherapy is given to a dose of 20 Gy in
five fractions or 30 Gy in 10 fractions.
and stridor
Chemotherapy is useful in chemosensitive Malignant central airway obstruction produces
338 tumours. Chemotherapy alone relieves SVCO in progressive symptoms of dyspnoea, stridor and
ONCOLOGIC EMERGENCIES 23
obstructive pneumonia. A central airway nar- cular weakness. If the concentration continues
rowing to <25% cross sectional area is usually to rise above 3.6 mol/l, neurological symptoms
needed to produce dyspnoea and stridor at rest. become more prevalent and patients may become
Airway obstruction can be produced by either confused, leading to coma and death.
extrinsic or intrinsic compression. Hypercalcaemia has few examination findings
Immediate measures are needed for sympto- specific to its diagnosis. However, a thorough
matic relief, which is monitored with pulse oxi- history and physical examination may help yield
metry. These include comfortable positioning, the underlying cause.
high-dose steroid (if known case of cancer, if not
withhold until definitive histologic diagnosis) Investigations
and supplemental oxygen or heliox (heliox is a
Along with serum corrected calcium, baseline
mixture of 80% helium with 20% oxygen; lower
laboratory investigations should include a full
density of helium helps in the easy flow through
blood count, urea, creatinine and electrolytes,
areas of turbulence and may decrease the work
liver function tests, serum phosphate, magnesium
of breathing). Muscle relaxants and respiratory
and PTH concentrations. ECG changes consist-
depressants should be avoided as are attempts
ent with hypercalcaemia include a shortened QT
to instrument airway without expert help and
and prolonged PR interval. At very high levels, a
positive pressure ventilation. CT scan of the
broad QRS complex may develop, T waves may
neck and chest may reveal the area and type of
flatten or invert and a variable degree of heart
obstruction.
block can occur.
Further management is shown in Figure
All further investigations are aimed at estab-
23.6. Stent results in prompt relief of symptoms;
lishing a cause and are likely to include imaging,
with cryotherapy 75% achieve symptomatic
particularly of the chest. In a patient without a
relief. Radiotherapy is given either as 20 Gy in
diagnosis of cancer, a myeloma screen should be
five fractions or 30 Gy in 10 fractions. Radio-
considered and in some patients a bone scan may
therapy response usually occurs within 72 hours
be helpful.
with 70–95% patients becoming symptom-free
by 2 weeks.
Management (Figure 23.7)
Saline rehydration is an important aspect of the
Malignancy associated management of hypercalcaemia. Hydration alone
hypercalcaemia may adequately treat mild hypercalcaemia.
Hypercalcaemic patients are universally de
Introduction
hydrated as a result of calcium-induced nephro-
Hypercalcaemia (a corrected serum calcium genic diabetes insipidus and poor oral intake.
concentration >2.6 mmol/l) occurs in up to The speed of administration of normal saline
30% of cancer patients during their illness. The depends on the degree of dehydration and renal
mechanisms of hypercalcaemia include secretion impairment, the level of hypercalcaemia and
of parathyroid-related-protein (80%), osteolytic the patient’s underlying cardiac function. Fluid
hypercalcaemia (20%) and rarely due to increased balance must be carefully monitored. The aim of
secretion of 1,25-D3 and ectopic secretion of saline treatment is two-fold, firstly, to increase
PTH. the glomerular filtration rate (GFR) and thereby
increase the filtered load of calcium into the
Clinical presentation tubular lumen from the glomerulus. Secondly,
The symptoms of hypercalcaemia are typically calcium reabsorption is inhibited in the proximal
non-specific and their severity depends on the nephron because of the calciuretic effects of
rapidity with which the calcium level rises. When saline. Once dehydration has been adequately
the serum calcium concentration increases above treated, loop diuretics can be used to increase
2.6 mmol/l, symptoms include fatigue, malaise, calcium excretion. Any drug that may be contrib-
depression, anorexia, nausea, vomiting, bone uting to the hypercalcaemia e.g. thiazide diuret-
pain, polydipsia, polyuria, constipation and mus- ics, lithium and calcium supplements should be 339
II SITE-SPECIFIC CANCER MANAGEMENT
Central airway
obstruction/stridor
• Stent/diathermy/laser Options:
• RT if not stentable • Stent
• RT if not stentable
Figure 23.6
Management of central airway obstruction/stridor.
discontinued. Hypophosphataemia is common erally reached at 7–10 days, with the beginnings
and phosphate levels should hence be monitored of a biochemical response evident at 2–4 days.
and replaced as appropriate. Up to 90% of patients will achieve normocalcae-
Bisphosphonates have been shown to be mia after a single dose; however, a second dose
superior to saline alone in the treatment can be given after 7–10 days if the calcium
of malignancy-associated hypercalcaemia. remains elevated. Patients with renal impairment
Bisphosphonates inhibit osteoclastic bone may require a dose reduction and this differs
resorption by adsorbing to the surface of bone between products. Although the administration
hydroxyapatite. The bisphosphonates of choice of bisphosphonates should not be delayed, prior
for the treatment of malignancy-associated rehydration may reduce the risk of further renal
hypercalcaemia are intravenous zoledronate, impairment. Another common adverse effect of
ibandronate and pamidronate. Pamidronate (60– intravenous bisphosphonates is a transient flu-
90 mg IV in 500 ml normal saline over 2 h) or like syndrome with fever, myalgias and chills.
Zoledronate (4 mg IV in 50 ml normal saline For patients in whom bisphosphonates are unsuc-
over 15 min) are the most widely used bisphos- cessful, alternative treatments such as glucocor-
phonates for this indication in the UK. The nadir ticoids, calcitonin and gallium nitrate can be
response to intravenous bisphosphonates is gen- tried following specialist endocrine advice.
340
ONCOLOGIC EMERGENCIES 23
Figure 23.7
Corrected serum calcium >2.6 mmol/l
Management of hypercalcaemia.
Options:
• Hydrocortisone 200 mg daily for 5 days
• Calcitonin 5 IU/kg s/c or IM 12 hourly
• Gallium nitrate 100–200 mg/m2 continuous IV for 5 days
• Dialysis – especially in patients with renal failure/resistant causes
Management
Management is essentially aimed at the prevention
of ATLS. It is important to identify patients at risk
of developing ATLS and to treat them prophylacti- allopurinol in reducing uric acid levels, but
cally with intravenous fluids and allopurinol. significantly more expensive.
Normal saline re-hydration should aim to Specific management of established ATLS
produce a urine output of approximately includes the close monitoring of uric acid, phos-
100 ml/h and diuretics may be required once phate, potassium, creatinine, calcium and LDH
euvolaemia has been achieved. Improving intra- levels. Fluid balance should be measured and
vascular volume, renal blood flow and glomeru- electrolyte abnormalities should be corrected
lar filtration rate promotes the excretion of uric (Table 23.2). Haemodialysis and intensive care
acid and phosphate. Alkalinization of the urine facilities should be available if required.
through the use of sodium bicarbonate is some-
times required. Hypersensitivity reactions
Allopurinol is a xanthine oxidase analogue,
which prevents the conversion of xanthine and Introduction
hypoxanthine to uric acid. It has proven efficacy Nearly all systemic agents used to treat cancer
in the prevention and treatment of hyperuricae- have the potential to cause hypersensitivity reac-
mia in patients with or at risk of ATLS. Allopu- tions; however, severe reactions are rare. Pro-
rinol prevents the formation of uric acid and vided patients receive appropriate pre-medication,
should be commenced prior to starting treatment close monitoring and prompt intervention when
with intensive chemotherapy. It has been associ- required, severe hypersensitivity reactions occur
ated with hypersensitivity reactions and reduces in less than 5% of patients. Platinum compounds,
the clearance of other purine-based chemothera- taxanes and monoclonal antibodies are most
peutic agents such as 6-mercaptopurine and likely to cause a reaction, although the timing of
azathioprine. such reactions may differ, as do their likely
Rasburicase is a recombinant urate oxidase underlying aetiology (Table 23.3).
that promotes the catabolism of uric acid to Platinum compounds such as cisplatin, carbo-
allantoin. Unlike allopurinol, there is no risk of platin and oxaliplatin classically cause reactions
xanthine nephropathy or calculi. It is licensed for following multiple cycles of treatment, typically
the treatment and prophylaxis of acute hyper after 6–8 doses. This is consistent with a type 1
uricaemia in patients who have high tumour hypersensitivity reaction following repeated
burden haematological malignancies and are at exposure to the drug and leads to IgE-mediated
high risk of ATLS. It is more effective than release of histamine, leukotrienes and prostag-
342
ONCOLOGIC EMERGENCIES 23
Table 23.2: Management of electrolyte Table 23.3: Showing the incidence of any
abnormalities grade hypersensitivity in a range of
chemotherapy agents
Electrolyte
abnormality Management Drug Incidence
Hyperphosphataemia Avoidance of additional Carboplatin or oxaliplatin 12–19%
phosphate
Paclitaxel 8–45%
Administration of a
phosphate binder e.g. Docetaxel 5–20%
calcium carbonate
Trastuzumab 40%
Haemodialysis/
haemofiltration Rituximab Up to
77%
Hypocalcaemia Calcium gluconate (10 ml
10% IV administered Cetuximab 16–19%
slowly with ECG
monitoring)
Hyperkalaemia Avoidance of additional
potassium
Cardiac monitoring less likely with each subsequent infusion. Delayed
Calcium resonium (15 g po reactions are still seen in 10–30% however, the
tds) underlying aetiology of these reactions is largely
Calcium gluconate
unknown.
(as above) for
cardio-protection
Insulin–dextrose (10–15
Clinical features
units insulin with 50 ml The National Cancer Institute Common Toxicity
50% dextrose) Criteria (NCI-CTC) distinguish between hy
Haemodialysis/
persensitivity and acute infusion reactions
haemofiltration
(Table 23.4).
Clinical features of hypersensitivity include
pruritis, rash, urticaria, rigors/chills/fevers, head-
ache, arthralgia/myalgia, tumour pain, fatigue,
landins from mast cells in the tissues and dizziness, sweating, nausea/vomiting, cough, dys-
basophils in the peripheral blood. This causes pnoea, bronchospasm, hypotension/hypertension
smooth muscle contraction and peripheral vaso and tachycardia.
dilatation, leading to urticaria, rash, angioedema,
bronchospasm and hypotension. Management
Although a similar clinical picture can be pro- Prophylaxis with antihistamines and corticoster-
duced following administration of a taxane, 95% oids is recommended for infusions that carry a
of these reactions occur during the first or second high risk of hypersensitivity reactions and for any
exposure to the drug and 80% occur within the patient that shows early signs of a hypersensitiv-
first 10 minutes of the infusion. In this scenario, ity reaction. Pre-medication for paclitaxel would
IgE-mediated type-1 hypersensitivity is unlikely, likely include 20 mg dexamethasone (preferably
and it is postulated that direct effects on mast given several hours before the infusion), chlor
cells and basophils lead to release of immu- pheniramine 10 mg and cimetidine 300 mg.
nomodulators and an anaphylactoid reaction. Patients should be closely monitored throughout
The solvent used in paclitaxel, but not docetaxel all infusions.
(Cremophor EL) has been shown to cause hista- Treatment of anaphylaxis is outlined in
mine release and hypotension and is felt to be Figure 23.8.
partly responsible for the hypersensitivity reac- Re-challenging patients following a hypersen-
tions seen with paclitaxel. sitivity reaction depends on the aim of treatment
Monoclonal antibody infusions can also and the severity of the reaction. The majority of
produce hypersensitivity reactions, which become patients who have a mild–moderate reaction
343
II SITE-SPECIFIC CANCER MANAGEMENT
Table 23.4: Grading of hypersensitivity reactions according to the NCI-CTC for adverse events
Grade
1 2 3 4 5
Hypersensitivity Transient Rash, flushing, Symptomatic Anaphylaxis Death
(allergic reaction) flushing or urticaria, dyspnoea, bronchospasm, with
rash, fever fever ≥38°C or without urticaria,
<38°C parenteral medication
indicated, allergy-
related oedema/
angioedema,
hypotension
Acute infusion Mild reaction, Requires therapy or Prolonged recurrence Life-threatening, Death
reaction infusion infusion interruption of symptoms following pressor or
(cytokine interruption but responds promptly initial improvement, ventilatory
mediated) not indicated, to symptomatic hospitalization support
intervention treatment, prophylactic indicated for other indicated
not indicated medication indicated clinical sequelae (e.g.
for ≥24 h renal impairment,
pulmonary infiltrates)
Secure airway and administer high flow oxygen usually less successful with approximately 50%
of patients experiencing recurrent hypersensitiv-
ity reactions.
Stop the infusion and cautiously recline the patient
Extravasation
Give 0.5 mg (0.5 ml of 1:1000) adrenalin IM
Repeat after 5 min if there is no clinical improvement Introduction
or there is further deterioration
An extravasation injury is any tissue damage that
occurs as a result of leakage of cytotoxic drugs
Give 10 mg Chlorpheniramine and 200 mg Hydrocortisone IV into the surrounding tissue.
• An irritant is a chemotherapeutic agent
Rapid infusion of IV fluids if required capable of producing venous pain at the ven-
Nebulized salbutamol if evidence of bronchoconstriction opuncture site or along a vein, with or without
an inflammatory reaction.
Figure 23.8
Management of anaphylaxis.
• A vesicant is an agent capable of blister for-
mation and/or tissue destruction.
Vesicant drugs are more likely to lead to
extravasation because of their potential for
during their first drug exposure (often seen with endothelial damage (Table 23.5). The risk of
taxanes and monoclonal antibodies) are likely to vesicant extravasation is very low, in the range
tolerate re-challenge of the drug if pre-medica- of 0.01–6% and although the use of implanted
tion is used with a slower rate of infusion. Desen- ports reduces the risk of extravasation, it can still
sitization protocols have been used with some occur.
success in patients who have a reaction to taxanes, Tissue damage occurs due to three main
however, re-challenge of platinum compounds is factors:
344
ONCOLOGIC EMERGENCIES 23
• Erythema, blotching, blistering, swelling and
Table 23.5: Vesicant vs non-vesicant drugs tenderness.
Non-vesicant/irritant/ • Early firm induration is often an indication of
Vesicant drugs exfoliant drugs
eventual ulceration.
Busulfan Bevacizumab • The skin may become white and cold, later
Carmustine Bleomycin developing into a black eschar.
Dactinomycin Carboplatin • Ulceration does not usually occur until at
(Actinomycin D) least 48 h, but may be delayed until one or
Daunorubicin Cisplatin
more weeks after the injury.
Doxorubicin Cyclophosphamide Management
Epirubicin Cytarabine Prevention is the key. The technique of cannula-
Mitomycin C Cetuximab tion and administration of the drug is important.
Treosulfan Docetaxel
Cannulas should ideally be inserted in the back of
the hand where they are readily accessible and
Vinblastine Etoposide
extravasation can be easily detected. Use of a
Vincristine Fludarabine central venous access device is preferable; however,
Vinorelbine Fluorouracil vesicant administration should only proceed
Gemcitabine
when there is blood return through the device.
Management of extravasation includes stop-
Ifosfamide
ping the infusion and aspirating from the intra-
Irinotecan vascular device. Extravasation of anthracyclines
Liposomal doxorubicin is initially managed with cooling of the area,
Methotrexate
however, heat application is generally used fol-
lowing the extravasation of plant alkaloids. Early
Oxaliplatin
review by the plastic surgeons for consideration
Paclitaxel of surgical debridement is vital when tissue
Rituximab necrosis is possible.
Topotecan
Hyaluronidase increases the permeability of
connective tissue and can be injected into the
Trastuzumab
extravasation sites of plant alkaloids. Used in
combination with a saline flush out of the area,
it aids the dispersion of the vesicant drug.
Dexrazoxane is a derivative of EDTA that
chelates iron. It has shown benefit in the treat-
1. Direct cellular toxicity of the antineoplastic ment of extravasation from intravenous anthra-
drug causing cellular necrosis. cycline therapy; although the exact mechanism
2. Differences in osmotic pressures between the of action is unknown. It is a systemic treatment
infused solution and the tissue environment. that is infused over 1–2 h each day for three days
3. Cellular damage due to the alkalinity or into a large vein, away from the extravasation
acidity of the solution. site. Side effects include nausea and vomiting,
diarrhoea, stomatitis, bone marrow suppression,
Clinical features elevated liver enzymes and infusion site burning.
Symptoms and signs of extravasation include:
Bone fractures or impending
• Pain, discomfort and burning sensations at or
around the cannula site.
fractures
• Reduced flow rate of the infusion. Bone metastasis is the third most common site of
• Lack of blood return through the cannula. metastatic disease. Bone metastasis can be lytic,
345
II SITE-SPECIFIC CANCER MANAGEMENT
347
Management of
common radiotherapy
side effects
TV Ajithkumar
24
General side effects Radiation reaction heals in 3–4 weeks fol-
lowed by tanning of the skin which takes a few
Skin more days to subside. Patients are asked to
Radiation dermatitis generally starts 10–14 days protect irradiated skin with sun block when sun
after the first fraction of radiotherapy and peaks exposure is unavoidable.
at the end or within one week of completion of Late skin reactions include atrophy and fibro-
radical treatment. It progresses from skin ery- sis of skin and telangiectasia. Radiation recall is
thema, to dry desquamation (dry, itchy, flaky), the phenomenon of skin reaction occurring in the
moist desquamation (raw painful area which previous radiation field when exposed to certain
may drain serous exudates) and finally necrosis chemotherapy (e.g. anthracyclines and gemcitab-
(rare). Skin reaction is intense when skin dose is ine). This is usually more severe than previous
high (e.g. electrons, bolus over the field), over radiation reaction and subsides in 2 weeks.
areas with skin folds (e.g. inframammary fold,
groin, perineum etc.), when radiotherapy is given Fatigue
concurrently with chemotherapy (particularly Fatigue (a subjective feeling of tiredness) is a
with anthracyclines, methotrexate, and 5-fluor- common side effect of radiotherapy and the exact
ouracil) and over areas of surgical wounds. cause is not known. Fatigue may increase pro-
Measures to minimize skin reaction include gressively during the radiotherapy and may take
avoiding mechanical, chemical and thermal irri- weeks or months to subside after radiotherapy
tations such as pat drying rather than rubbing, treatment. Measures to improve fatigue include
using simple soap, avoiding perfumes, powders, frequent periods of rest, regular minimal aerobic
deodorants over the irradiated area, shaving with exercise, and stress management. A study showed
razor blade, heat, cold and sun and using loose that 20–30 minutes of brisk walking 4–5 times
fitting and cotton clothes. 1% hydrocortisone per week improved fatigue level, symptom inten-
cream is useful for itchy areas. sity and physical functioning.
During dry desquamation, hydrophilic mois-
turizing ointment with no heavy metals (e.g. Anorexia
aqueous cream) can be applied to the skin after Some patients experience anorexia and the exact
radiation treatment every day. For moist desqua- cause is not known. A number of factors such as
mation, dress the moist area with hydrogel, the primary cancer itself and its systemic treat-
hydrocolloid or alignate dressing and continue ment as well as concurrent medications can influ-
applying aqueous cream to other areas. When ence the degree of anorexia. Measures to improve
radiotherapy is stopped, zinc oxide or silver sul- anorexia include frequent small meals, high
fadiazine (flamazine) may be applied to the skin energy high protein diet, and sometimes appetite
with non-adhesive dressing. Superadded infec- stimulants like megestrol acetate (400–800 mg
tion may need antibiotics depending on culture daily) or steroids. Anorexia can cause clinically
and sensitivity. significant weight loss (loss of ≥10% prediagnosis
348 © 2011, Elsevier Ltd
DOI: 10.1016/B978-0-7234-3458-0.00029-4
MANAGEMENT OF COMMON RADIOTHERAPY SIDE EFFECTS 24
weight loss) in which case enteral feeding is • Mouth wash – sterile water, 0.9% saline
advised, particularly to those receiving radical or sodium bicarbonate solutions. Normal
radiotherapy. saline can be prepared by adding one tea-
spoon of salt to 1 litre of water and 1–2
Bone marrow suppression tablespoons of sodium bicarbonate can be
Irradiation of a large area of marrow bearing added if the saliva is viscous.
bones can result in bone marrow suppression. • Benzydamine (Difflam) is recommended
The degree of bone marrow suppression is high for prevention of oral mucositis whereas
if concurrent myelosuppressive chemotherapy is chlorhexidine, sucralfate and antimicro-
used. All patients at risk of bone marrow sup- bial lozenges are not.
pression need weekly blood counts and appropri- • Pain – controlled by topical anaesthetics and
ate action. analgesics and systemic analgesics.
• Diet – soft bland diet and in severe mucositis,
enteral feeding.
Site-specific side effects
Xerostomia
Brain
During radiotherapy, dry mouth starts within
Cerebral oedema 1–2 weeks of radiotherapy which involves the
Radiotherapy can cause cerebral oedema which region of salivary glands. Later saliva becomes
manifests as headache, vomiting, seizures, neuro- thick and sticky which causes problems of retch-
logical deficit and altered mental function. Ster- ing, coughing, nausea and vomiting, difficulty in
oids are the treatment of choice. talking and disturbed sleep. Dry mouth can
become chronic. Measures include:
Alopecia
• Mouth care – frequent cleansing.
Alopecia depends on the dose and extent of radi- • Improve moisture – frequent sips of fluids and
ation. Alopecia starts when the radiation dose is saliva substitutes.
above 25–30 Gy and is permanent when the dose • Stimulate saliva – sucking candies, pharmaco-
is >40 Gy. If regrowth occurs it usually starts 2–3 logical measures (e.g. oral pilocarpine 5 mg
months after completion of radiotherapy. 3–4 times daily).
• In suitable patients, intensity modulated radi-
Head and neck cancer otherapy (IMRT) delivery (p. 85) can avoid
Mucositis irradiation of at least one salivary gland and
The initial reaction is tenderness in the mouth thereby reduce the degree of xerostomia.
with associated erythema and oedema of the Taste change
mucosa. Subsequently a whitish membrane is
formed, which progresses to a painful ulcer with Radiation can change taste sensation, which may
continuation of radiotherapy. Bleeding and persist for years. It usually starts after doses
superadded infection can occur. Concurrent >20 Gy and around 90% patients receiving
chemotherapy and metallic fillings in teeth (by radical radiotherapy are affected. Measures
electron back scatter) can enhance mucosal skin include mouth care before and after meals and
reaction. Dental assessment prior to radiother- modification of food and preparation. An
apy is important (p. 79). Measures to prevent unpleasant taste of meat is one of the common
and manage mucositis include: problems, which can be masked by trying addi-
tional seasoning, marinating meat in wine or
• Radiotherapy planning – use of midline radia- sweet and sour sauce before and during cooking
tion blocks wherever appropriate and 3D and serving food cold or at room temperature.
planning reduce mucosal injury.
• Mouth care – frequent cleansing, tooth Hypopituitarism and hypothyroidism
sponges and swabbing rather than use of a Patients who received radiotherapy to the pitui-
toothbrush. tary and thyroid regions are at risk of hormonal
349
II SITE-SPECIFIC CANCER MANAGEMENT
insufficiency. Patients need hormone replacement needed. Dietary modifications with low fat, low
treatment. sugar diet and high fluid intake are also useful.
Thoracic region Diarrhoea
Diarrhoea can occur 2–3 weeks after starting
Cough
treatment. The pattern of diarrhoea may be
Cough is initially productive which becomes dry either increased frequency of stool or watery
as treatment progresses. Treatment is sympto- stools with abdominal cramps. Sulfasalazine
matic with adequate fluid intake, humidification 500 mg twice orally is suggested to reduce the
of air, cessation of smoking, cough suppressants incidence and severity of radiation-induced enter-
and antibiotics if there is superadded infection. opathy in patients receiving pelvic radiotherapy.
Pneumonitis A low-residue, low fat (<40 g) diet and avoid-
ance of milk products can be useful in reducing
Acute pneumonitis usually occurs 1–3 months
diarrhoea. Antidiarrhoeal medications such as
after radiotherapy and rarely earlier (‘hyper
codeine and loperamide are useful when dietary
acute’). It manifests initially with dry cough and
modifications are ineffective.
later with productive cough, fever and dyspnoea.
Treatment includes bed rest, steroids (pred- Proctitis
nisolone 1 mg/kg for 4–6 weeks) and antibiotics Patients with tenesmus benefit from antispas-
if superadded infection. modics and anticholinergics. Pain can be control-
led by frequent sitz baths and local creams
Oesophagitis such as haemorrhoidal preparations. Patients
Oesophagitis develops 2–3 weeks after starting with chronic proctitis are treated with sucralfate
radiotherapy and the severity is high with con- enema.
current chemotherapy, long length of oesopha-
gus in the radiation field and increased dose per Cystitis
fraction. Patients develop progressive painful Cystitis manifests with dysuria, nocturia, urgency,
swallowing. Measures to improve dysphagia hesitancy and frequency. Patients are encouraged
include liquid high calorie, high protein diet, to have an adequate fluid intake. Bladder infec-
local anaesthetics, liquid analgesics and systemic tion should be treated. Bladder analgesics (e.g.
analgesics. Patients may develop superadded can- phenazopyridine) and antispasmodics are useful
didiasis and need antifungal treatment. in pain. Patients receiving prostatic radiotherapy
can develop bladder outflow irritation which
Radiation fibrosis manifests with hesitancy, decreased force of flow
Radiation fibrosis develops 6–12 months after and frequency. In these situations, alpha blockers
completion of radiotherapy and can manifest such as terazocin (Hydrin) and tamsulosin
with dyspnoea. Treatment is essentially symp (Flomax) result in better symptom control than
tomatic. NSAIDs.
Vaginal stenosis and dryness
Abdomen and pelvis
Vaginal stenosis and dryness are late effects.
Vomiting Treatment is symptomatic with lubricants and
Vomiting is common with radiation involving vaginal dilatators.
the upper abdomen. Vomiting can occur within
Erectile dysfunction
6 hours of radiotherapy and may last for 3–6
Erectile dysfunction results from damage of
hours. Patients receiving treatment to the upper
pelvic nerves and fibrosis of pelvic vasculature.
abdomen, large single fractions and total body
Pharmacological interventions, devices and pros-
or lower hemibody irradiation are advised to
thesis are useful.
have prophylactic anti-emetics with a serotonin
receptor antagonist (e.g. ondansetron 8 mg, 30 Gonadal failure
minutes prior to radiotherapy). In severe vomit- Ovarian failure can induce symptoms of meno-
ing continual administration of antiemetics is pause and permanent sterility (>8 Gy fractionated
350
MANAGEMENT OF COMMON RADIOTHERAPY SIDE EFFECTS 24
dose). HRT is useful if not contraindicated. In appropriate the testes should be shielded from
young people needing pelvic radiotherapy, laparo- radiation.
scopic oophoropexy (surgically placing ovaries
outside radiation field), may be protective. Further reading
Testicular irradiation (>12 Gy of fractionated Faithfull S, Wells M (ed). Supportive Care in Radiotherapy.
RT) causes permanent sterility and hence if Edinburgh: Churchill Livingstone, 2004.
351
Clinical trials in cancer
TV Ajithkumar and HM Hatcher
25
Introduction Preclinical stage
Research and development is an important aspect The preclinical stage involves screening of poten-
of cancer management. Cancer drugs have to tial agents and their testing on animal models.
undergo rigorous evaluation before approval for Initial testing may be performed on cell lines or
clinical use. However, technological develop- tumour explants before moving to an animal
ments, such as a new surgical or radiotherapy model. Preclinical studies establish that the
technique, often do not undergo such complex potential agent is not lethal, safe to use and that
procedures. They are approved if they are proven there is some indication of activity in the medical
not to be harmful and they meet the appropriate condition studied.
quality control measures. In fact, many of the
new radiotherapy techniques are approved for
clinical use without any robust comparative data Clinical trials
on the clinical efficacy.
Traditionally the clinical efficacy of drugs is Phase I
studied through several phases of clinical trial Phase I studies are the first human application of
after initial pre clinical studies. Each of these a new drug or drug combination. The aim of this
trials is aimed at generating sufficient evidence phase is to establish the dose and schedule of the
on the safety, dosing, efficacy and feasibility of experimental agent for efficacy testing in a phase
use. This process is expensive and time consum- II study based on the maximum tolerated dose
ing (Figure 25.1), which often leads to very (MTD) of the new agent. Phase I studies are typi-
high costs of effective treatments preventing cally single arm, open label, sequential studies
their general use and often prompting policy that include patients with good performance
makers to adopt rationing strategies based on status (PS 0-2) and for whom there is no standard
cost-effectiveness. treatment option. The MTD is determined by
New methods of trial design are constantly progressively increasing the dose in small cohorts
evolving, both to address the length of time to until the dose limiting toxicity (DLT) is achieved.
take a drug from development into practice and DLT is defined as grade 4 non-haematological
to answer questions which apply to newer drugs. toxicity or a grade 3 or more haematological
For example targeted treatments may incorpo- toxicity graded according to the National Cancer
rate an assessment of the efficiency in affecting Institute Common Terminology Criteria for
that target or in measuring the sub-population Adverse Events (CTCAE) (p. 38). MTD dose is
who may benefit most from an effective but defined as the dose below the level the DLT is
expensive treatment. met or the dose level at which the DLT is seen.
Figure 25.1
Concept and discovery Drug approval, Process of drug discovery.
Pre-clinical studies Clinical trials
launch and sale
Synthesis Registration
Research of active Screening Preclinical Preclinical Phase I Phase II Phase III with drug Production
target substance trials I trials II studies studies studies authority and sale
PRE-DISCOVERY
There are a number of components to the design Box 25.1: Components of phase I study design
of a phase I study and there are various dose and methods of dose escalation
escalation methods to find out the maximum tol- Design components of a phase I study
erated dose (Box 25.1). Phase I trial design is a • Starting dose – usually 110 of the animal dose
specialized topic and is beyond the scope of this • Dose increment
chapter (see Further reading). • Dose escalation method
• Number of patients per dose level
• Specification of dose-limiting-toxicity (DLT)
Phase II • Target toxicity level
Phase II studies are done to assess the activity, • Definition of maximum tolerated dose (MTD)
safety and feasibility of new drug. There are • Recommended dose and patient selection for
many options for the design of a phase II trial. phase II trial
A phase II study can either be completed in a Dose escalation methods
single stage or as two stages. In two stage study, • Rule-based design – based on no prior assumption
of the dose-toxicity curve and allows dose
the first stage is to evaluate the treatment efficacy escalation and de-escalation of dose with
(phase IIA) and in the second stage (phase IIB) is diminishing fractions of preceding dose depending
to identify promising agents to send to phase III on the absence or presence of severe toxicity in
the previous cohort of patients treated.
for additional evaluation. Phase IIA design is to
• 3 + 3 design – most frequently used method.
test short-term tumour response, whereas phase The classical assumption is toxicity increases
IIB design compares two or more regimes which with dose. It starts with a cohort of three
have demonstrated some initial efficacy. patients and subsequent cohorts are treated at
increasing level.
Randomized phase II trials are increasingly
• Accelerated titration design
being used in cancer. These studies randomize
• Pharmacologically guided dose escalation
patients to new treatment or standard treatment
• Model based designs – are complex and use
or to a number of new treatments. Randomi statistical models that actively seek a dose level
zation is to ensure a similar patient population that produces a pre-specified probability of dose
in each group and not to do a statistical test to toxicity using dose-toxicity curve calculated from
all enrolled patients. Examples include:
compare treatment groups. These trials are not
• Continual reassessment method
adequately powered to do this and this is the
• Escalation with overdose control
purpose of a phase III trial. Treatment outcomes
• Designs that use time-to-event endpoints
are ranked according to activity, safety and fea-
• Designs that use toxicity and efficacy as
sibility to take the drug forward to phase III endpoints
study.
356
CLINICAL TRIALS IN CANCER 25
357
III RESEARCH IN ONCOLOGY
358
CLINICAL TRIALS IN CANCER 25
Further reading
Target modulation No target modulation Phase I
(30 patients) Le Tourneau C, Lee JJ, Siu LL, Dose escalation methods
in phase I cancer clinical trials. J Natl Cancer Inst
2009;101(10):708–720.
Phase I/II No further Kummar S, Doroshow JH, Tomaszewski et al. Phase 0
Dose escalation and studies Phase II clinical trials: recommendations from the Task Force on
efficacy and target (300 patients) Methodology for the Development of Innovative Cancer
assessment (around
Therapies. Eur J Cancer 2009;45(5):741–746.
100 patients)
Eisenhauer EA, O’Dwyer PJ, Christian M, Humphrey JS.
Phase I clinical trial design in cancer drug development.
Phase III
(3000 patients) J Clin Oncol 2000;18:684–692.
Phase II/III Lee JJ, Feng L. Randomized phase II designs in cancer
efficacy and target clinical trials: current status and future directions. J Clin
qualification (around Oncol 2005;23(19):4450–4457.
1000 patients) Green S, Bendetti J, Crowley J. Clinical Trials in Oncology.
Second edition. London: Chapman and Hall, 2003.
Eisenhauer EA, Twelves C, Buyse M. Phase I cancer clinical
trials. Oxford: Oxford University Press, 2006.
Drug approval
Figure 25.2
Phases of the clinical trials.
359
26
Understanding the
strengths and
weaknesses of clinical
research in cancer
C Williams
‘I hope that the cares of medical practice What are the important
have not obliterated your interest on
solving deductive problems.’ elements when assessing
Sherlock Holmes to Watson in the reports of trials?
Stockbroker’s Clerk* By far the most important elements are the design
and conduct of the trial. The methods of analysis,
Introduction while still important, are generally less likely to
lead to inappropriate conclusions than inappro-
This chapter briefly discusses cancer clinical trials priate design and management of a trial. This
and systematic reviews of the cancer literature, chapter is based on the methods recommended
together with the concept of critical appraisal by various EBM groups (Box 26.1).
and evidence-based medicine (EBM). Much of Because there is such a huge volume of medical
literature appraisal requires a common sense, but literature published each year, it is important to
systematic, approach to ascertaining answers to take a systematic approach to what you appraise
key questions about potential biases in the design, thoroughly (Box 26.2). The first step in appraisal
conduct and reporting of a study or review. is to screen the paper to see if it is worthy of
careful reading. It may be possible to answer
Appraising reports of these screening questions on reading the title and
abstract.
cancer clinical trials
The difficulty in appraising clinical trials lies in Step 1 – Screening questions
assessing the strength of the evidence. On cursory • Was there a clearly focused question? Impor-
reading the author of the report may seem to tant aspects include the population studied,
present a strong case for the trial, but you need the interventions and the outcomes used.
to see beyond the rhetoric, the eminence of the • Did the investigators use the right type of
author or institution/trials group and the status study design for the question being asked? If
of the journal publishing the paper, to assess the there is a clearly focused question that
evidence and to see if it is strong enough to stand is addressed by a trial of appropriate design
on its own. Very strong evidence may be enough it is worth continuing on to a full appraisal
to influence practice by itself, but more com- of the results. However, do not save time
monly the evidence is weaker and needs support by simply reading the abstract (Example
from further studies. Box 26.1).
Step 2 – Appraising a paper
*Quotes in this chapter are as those cleverly used by Jorgen reporting a trial
Nordenstrom in his book ‘Evidence based medicine in Sherlock
Holmes’ Footsteps’ (Wiley Black-well, 2006). There are a number of crucial questions that need
to be answered (Box 26.2):
360 © 2011, Elsevier Ltd
DOI: 10.1016/B978-0-7234-3458-0.00031-2
UNDERSTANDING THE STRENGTHS AND WEAKNESSES OF CLINICAL RESEARCH IN CANCER 26
number is not an acceptable method, though it is life are often of major importance; they are fre-
sometimes referred to as pseudo-randomization. quently ignored in the primary research.
Since the researchers know which treatment each
person is allocated to before they consent, selec- Optimal cut points and the problem with
tive allocation can occur, which will skew the multiple comparisons.
results. Hence the process used for randomization ‘While the individual man is an insoluble
must ensure that neither the trial subjects nor the problem, in aggregate he becomes a
investigator can influence the treatment arm each mathematical certainty. For example you
person ends up in (‘allocation concealment’). can never foretell what any one man will
Despite their pre-eminence, RCTs do, however, do, but you can say with some precision
have their limitations. They are usually carried what an average number will be up to.
out in a narrowly defined research setting. Pro- Individuals vary, but percentages remain
tocols require careful selection of patients with constant.’
specific characteristics. There may be a propor- Sherlock Holmes, in The Sign of The Four
tion of patient eligible who chooses not to par-
ticipate. Because of these factors, patients in an Where there is a continuous outcome measure, it
RCT may not be typical of patients in routine is common to simplify analysis by defining one
health practice. or more points that are used to characterize risk.
The way that these points are selected may have
2. Was the study based on a major effect on the analysis.
a pre-specified protocol?
Subgroup analysis
A protocol written before the trial starts is a
Subgroup analyses are fairly common in trials;
prerequisite to good research and some journals
they use the data from a study to compare one
are now recommending that the original protocol
endpoint across different subgroups. There are
be submitted with the final paper to ensure that
three major problems with this approach:
there was such a protocol and to identify any
deviations from the original design. Any devia- • The comparison is not randomized.
tion from the original study design can result in • The comparison is less precise as there are
skewed observation of clinical benefit. numbers of patients and those reaching end
When reading a paper bear in mind the fol- points is smaller.
lowing elements of trial design: • The power calculations for the size in a study
may be invalidated by a large number of
Was there a clearly defined research question?
subgroups.
A good study has limited objectives that are
based on a biological hypothesis. Studies with no Where a subgroup is found to behave differ-
focus and without a clearly defined question are ently, you should consider if there is a plausible
in danger of becoming a ‘fishing expedition’ with biological mechanism for this and whether other
multiple comparisons. trials have found a similar finding. Where there
The chances of multiple testing increases when is a statistical analysis, this should be done as a
there is numerous subgroups or endpoints or formal test of interaction. Strong empirical evi-
when data are reported on multiple side effects. dence suggests that post hoc subgroup analyses
This is particularly problematic when such com- often lead to false positive results (Example Box
parisons are unplanned. 26.2).
Did the study have appropriate end points?
Most RCTs in cancer therapy concentrate on 3. Did the investigators stick to the protocol?
reporting response rates (a surrogate end point Clinical research is rarely predictable, so changes
of clinical benefit), disease free survival and or alterations to a protocol are often needed.
overall survival. However, accepting that cure is However, they should be clearly reported, as
out of the reach of many patients with cancer, major deviations may reduce the reliability of
information on symptom control and quality of the data.
362
UNDERSTANDING THE STRENGTHS AND WEAKNESSES OF CLINICAL RESEARCH IN CANCER 26
Box 26.4: Is the result important? Example Box 26.4: Publication bias
In any study, the beneficial effect/risk of the Stern JM, Simes RJ. Publication bias: evidence of
experimental arm can be due to three possible delayed publication in a cohort study of clinical
reasons: research projects. BMJ 1997;315:640–645.
• Bias – excluded by assessing the validity of study. This retrospective study was undertaken to determine
• Chance variation between the treatment groups the extent to which publication is influenced by study
– excluded by analysing p-value and confidence outcome. Of the 218 studies analysed with tests of
interval. significance, those with positive results (p < 0.05)
• The real benefit/risk – quantified by calculating were much more likely to be published than those
various event rates. with negative results (P ≥ 0.10) (hazard ratio 2.32
(95% confidence interval 1.47 to 3.66), p = 0.0003),
P-value is used to measure the probability of with a significantly shorter time to publication
occurring the benefit/risk by a chance, e.g. a p-value (median 4.8 vs. 8.0 years). Studies with indefinite
of 0.01 means that there is a 1 in 100 (1%) conclusions (0.05 ≤ P < 0.10) tended to have an even
probability of the result occurring by chance. lower publication rate and longer time to publication
Conventionally, a p value of <0.05 (<1 in 20 than studies with negative results (hazard ratio 0.39
probability) is set as the statistically significant result. (0.13 to 1.12), p = 0.08).
Confidence interval (CI) is used to measure the
sampling error. Since any study can only examine a
sample of a population, we would expect the sample
to be different from the population (sampling error). ing the results of a meta-analysis, you should ask
Conventionally a 95% CI is used, which specifies that the following questions:
there is a 95% chance that the population’s true
value lies between the two limits. If the 95% CI • Were all the available studies included? An
crosses the ‘line of no difference’ between
interventions, the result is not statistically significant.
incomplete search of the literature can bias
Once the bias and chance have been ruled out, the
the findings of a meta-analysis or systematic
benefit/risk of intervention can be quantified using review.
the following measures: • Was it appropriate to pool the data? Hetero-
• Relative risk (RR) – the ratio of the risk in the geneity among studies may make any pooled
experimental group divided by the risk in the
control group.
estimate meaningless.
• Absolute risk reduction (ARR) – the difference
• Was the quality of the trials good? The quality
between the control event rate and experimental of a meta-analysis cannot be any better than
event rate. This is a more clinically relevant the quality of the studies it is summarizing.
measure.
• Relative risk reduction (RRR) – calculated by Did the reviewers find all of the trials?
dividing ARR with control event rate. One of the greatest risks of bias in a meta-
• Number needed to treat (NNT) – is the inverse of
ARR and is the most useful measure of clinical
analysis is omitting relevant studies. Often studies
benefit. It tells you the absolute number of are never published; evidence suggests that such
patients who need to be treated to prevent one studies are likely to be negative. This is known
adverse outcome.
as publication bias (Example Box 26.4). Regis-
tration of trials, before they start, is being intro-
duced as a way of avoiding publication bias.
width of the confidence intervals. If there is an
inadequate sample size, the confidence intervals Duplicate publication
will be abnormally wide. Studies which are positive are more likely to
appear more than once in publication. This is
Systematic reviews especially problematic for multi-centre trials
where an individual centres may publish results
and meta-analyses specific to their site.
Introduction The limitations of a Medline search
A systematic review is one carried out in While a Medline search is the most convenient
accordance with a written protocol and using way to identify published research, it should not
methodology designed to reduce the risk of bias. be the only source of publications for a meta-
Meta-analysis is the quantitative pooling of data analysis. Medline searches cover only about a
from two or more studies. When you are examin- quarter of the medical literature.
366
UNDERSTANDING THE STRENGTHS AND WEAKNESSES OF CLINICAL RESEARCH IN CANCER 26
Foreign language publications
Box 26.5: NICE appraisal on addition of
Some meta-analyses restrict their attention to paclitaxel to platinum in ovarian cancer
English language publications only. This makes ‘While design differences between the four trials,
life easier for the reviewers, but there is evidence in terms of severity of disease of included patients,
that researchers tend to publish reports of posi- differences in treatment and control drugs and
doses, length of follow-up, and the extent of
tive trials in English language journals. Con- cross-over (before and after disease progression),
versely, they publish negative reports in a native may hamper statistical pooling of results, meta-
language journal where the citation index is analyses have been undertaken … These take
account of statistical heterogeneity as far as
likely to be lower. possible, and their results appear consistent,
reporting that the findings for progression-free
Heterogeneity survival (hazard ratios = 0.84, 95% CI = 0.70 to 1.02
[MRC] and 0.87, 95% CI 0.72 to 1.05 [BMS]) and
Even when the trials do not have obvious clinical overall survival (hazard ratios = 0.82, 95% CI 0.66
heterogeneity the results may turn out to be very to 1.01 [MRC] and 0.82, 95% CI 0.68 to 1.00 [BMS])
different. An example is RCTs testing the efficacy across the trials do not show statistically significant
differences between paclitaxel/platinum and the
of adding paclitaxel to platinum therapy for the alternatives.’
primary chemotherapy of ovarian cancer. The ‘The four trials showed consistently that treatment
initial two studies, GOG 111 and OV10, showed with paclitaxel in combination with platinum leads to
an advantage for the addition of paclitaxel. Two more side effects …’
subsequent trials, GOG132 and ICON3 (by far ‘The Committee took account of this range of trial
evidence as well as other factors that would
the biggest trial), have failed to show an advan- differentiate between the two regimens including
tage for the addition of paclitaxel. Examining the the side-effect profiles of the treatments … On this
results there appears to be such an extreme het- basis the Committee considered that paclitaxel/
platinum combination treatment should no longer be
erogeneity that pooling of the data in a meta- recommended exclusively as standard therapy for
analysis should be avoided. Despite this NICE women receiving first-line chemotherapy for ovarian
in its appraisal did include a meta-analysis cancer. [The original recommendation was made
when ICON3 had not been published.] … both
(Box 26.5). platinum therapy alone and a combination of
The main areas where heterogeneity occurs paclitaxel and a platinum compound were
include: appropriate first-line treatments for women with
ovarian cancer.’
Heterogeneity of the treatment and control This guidance allows clinicians to exercise their
groups: prejudice, either to believe the two positive trials or
the two negative trials, containing many more
• Varying inclusion and exclusion criteria. patients. The meta-analysis, although showing no
• Variation in general health of patients or statistical benefit for the addition of paclitaxel, has
geographical differences. not added to the guidance since its conclusion has
been ignored. The statistical heterogeneity was
• Differences in dose or timing of a drug or so great that it would have been better to have
co-medication. discussed the two pairs of trials in narrative fashion,
Heterogeneity in the design of the study: rather than to have produced a meta-analysis of the
four trials.
• Variation in length of follow-up.
• Variation in proportion of drop outs and
how they are handled.
Heterogeneity in the management of the jects, increases confidence in that treatment.
patients and in the outcome: Where there is excessive heterogeneity pooling
• Variation in the management of of data should be avoided. The results can be
co-morbidity. explored in various ways.
• Variation in managing side effects.
Subgroup analysis
Managing heterogeneity When there is substantial heterogeneity, you can
A degree of heterogeneity is to be expected and look and compare subgroups of the studies. As
is acceptable. Showing that a treatment gives the case in Box 26.5, of the use of paclitaxel
consistent results, with some variation, across a chemotherapy in ovarian cancer, suggests rather
variety of trials, and in a large number of sub- than pooling all the data, the review could have
367
III RESEARCH IN ONCOLOGY
pooled the data from the two groups of positive number of positive studies exceeds the negative
and negative trials and then discussed the reasons group or vice versa. However, there is a major
for the different outcomes. flaw with this approach as it ignores the possibil-
ity that some studies are negative because they
Meta-regression are simply too small.
You can try to adjust for heterogeneity in a meta-
analysis. This works similarly to the adjustment How to avoid bias from exclusion
for covariates in a regression model. of publications
The major bibliographic databases, registries for
Sensitivity analysis clinical trials, and the grey literature should all
Another approach to heterogeneity is to examine be searched. In addition the bibliographies of all
the sensitivity of the results by looking at selected articles found should be checked to see if there
groups of studies. This may mean only including are relevant trial reports.
those trials selected to be of good quality factor
or some other element likely to reduce bias. Intentional exclusion of studies
In any meta-analysis, you have to draw a line
Was the quality of the trials good?
somewhere. Studies that fail to meet your criteria
Meta-analysis cannot correct or compensate for will not be included in the results. Where the
poorly designed or conducted studies. It is for cut-off point is based on judgment, such as trial
this reason that poor quality trials should be quality, this can sometimes cause serious contro-
excluded or a sensitivity analysis carried out versy. A Cochrane Review of mammographic
based on trial quality. screening for breast cancer found seven eligible
Reporting of trials is often poor so that it is studies, but only two were deemed to be of suf-
impossible to assess the quality of the study. ficient quality to include in the review; meta-
There is evidence that poorly reported trials are analysis of the two studies found no benefit from
more likely to be of poor quality and incomplete screening. If all seven studies were included the
reporting is associated with poor quality. outcome was positive. This Cochrane review
provoked a furious response and intense debate
Taking the easy option – over the rights and wrongs of excluding most of
a narrative review the available trials (Example Box 26.5).
‘The temptation to form premature
theories upon insufficient data is the bane Objectivity in a systematic review
of our profession.’ ‘It is capital mistake to theorize before one
Sherlock Holmes in the Tragedy has data. One begins to twist the facts to
of Birlstone fit theories, instead of theories to fit facts.’
Sherlock Holmes in Scandal in Bohemia
A classical narrative review frequently suffers
from a lack of systematic ways of avoiding bias. A systematic review or meta-analysis should have
There is also a worrying tendency to choose a written protocol, though most published meta-
papers that support the reviewer’s own point of analyses do not state if a protocol was used. This
view. This is fine in an opinion piece designed to protocol should state: the inclusion/exclusion
make people think, but is very dangerous if the criteria for studies, how quality will be assessed,
reviewer purports to give a balanced assessment a detailed search strategy and the statistical
of the literature. methods used to combine results. The protocol
Traditional narrative reviews generally do not is there to reduce bias, increase transparency and
use meta-analysis, but will often undertake an to make it possible to see how the review was
exercise often referred to as vote counting. In done. As well as including information on
such a process, there is a sum of the number of included studies, a systematic review should
positive and negative studies; the overall inter- report on which studies were excluded and on
pretation depending on whether or not the what grounds.
368
UNDERSTANDING THE STRENGTHS AND WEAKNESSES OF CLINICAL RESEARCH IN CANCER 26
369
Appendix:
A glossary of common targets
with examples
HM Hatcher
Figure A.1
EGFR EGFR receptor showing the extracellular binding domain (pink)
and intracellular tyrosine kinases (blue). When the EGF (ligand)
(purple) binds the receptor it causes heterodimerization and
EGF autophopsphorylation (yellow P symbols) of the tyrosine kinase
domains within the cell.
Extracellular domain
Intracellular domain
P P
P P
EGF EGF-R
Extracellular domain
Intracellular domain
P P
DAG PIP2 PLCα P P Sos-1 Ras
Grb2
PI3K Shc
Raf MEKK-1
IP3 JAK1
Ca2+
Transcription Nucleus
factors ELK-1 ELK-1
C-jun
STAT 3 STAT 1
STAT 3 STAT 3
Figure A.2
The complex signalling pathways following activation of EGFR.
371
APPENDIX: A GLOSSARY OF COMMON TARGETS WITH EXAMPLES
Intracellular domain
Figure A.4
The effect of angiogenesis on
tumour growth (above) and the
effect of inhibiting angiogenesis
Tumour (below).
Angiogenesis
Tumour growth
Blood supply
with those who have specific EGFR mutations. VEGF. VEGF is another growth factor receptor
Transtuzumab (Herceptin™) is an antibody which, like EGFR, leads to an intracellular cascade
against HER-2 and has shown significant activity of proteins which regulate cell growth. Increased
in breast cancer. In the adjuvant setting, com- VEGF expression is also observed in many cancers
bined with chemotherapy, it has shown signifi- (e.g. ovarian and renal). Interfering with this
cant improvements in progression free survival process by reducing VEGF expression or its sub-
(p. 128). Lapatinib is a small molecule which has sequent signalling has been another major target
activity against both HER-2 and EGFR and is of drug developers (Figures A.4 and A.5).
also active in breast cancer. As with EGFR, antibodies can be developed
to the receptor and bevacizumab is a VEGFR
Vascular endothelial growth antibody which has shown activity in breast,
colorectal and renal cancer and investigations
factor (VEGF) are underway for other malignancies. TKIs which
Angiogenesis is an important process by which work on this pathway include sorafenib and
tumours gain increased vasculature allowing sunitinib both of which have shown significant
them to grow and spread. A major contributory improvements in survival in metastatic renal
factor to angiogenesis is increased expression of cancer (Figure A.6).
372
APPENDIX: A GLOSSARY OF COMMON TARGETS WITH EXAMPLES
Figure A.5
Potential downstream effects of
Vascular endothelial VEGF VEGFR-2 VEGF.
cell plasma membrane
Extracellular domain
Intracellular domain
P13K P P Raf
P P MEK
Figure A.6
Sites of action of various drugs
which work in the VEGF/
VEGF-receptor 2 angiogenesis pathway.
Bevacizumab VEGF
Extracellular domain
Intracellular domain
PKC
Sunitinib
Sorafenib
SPK Ras
P13K Raf
Sorafenib
Akt/PKB MEK
ErK
Angiogenesis
inhibitors and PI3K inhibitors which are under the bcl-2 antisense Oblimersen sodium which in
investigation. The mTOR inhibitor Terosilimus early trials has shown activity in pre-clinical and
has already shown activity in metastatic renal pilot studies of lung cancer and melanoma.
cancer. Future areas involve targeting many parts of
Antisense agents are those which recognize the signalling pathway at the same time to see if
abnormal proteins or products of transcription, this leads to synergistic activity and is safe.
bind to them and inactivate them. An example is
374
Index
Antiretroviral therapy (ART), 295 Basal cell carcinoma (BCC), Blumer’s shelf, 142
see also highly active cutaneous, 237–239, Body image, children and young
antiretroviral therapy 238b–239b, 238f adults, 326
Antisense agents, 374 Basal naevus syndrome, 264 Bone
APC gene mutations, 52 BCG, intravesical, 178–179 fractures/impending fractures,
Apocrine carcinoma, 244 Bcl-2 antisense molecule, 314, 374 345–346, 346f, 346t
Aromatase inhibitors (AI) BCR-ABL1 fusion gene, 311–312 late effects of cancer treatment,
breast cancer, 127–128 BEAM chemotherapy, 305t 61–62, 61b
endometrial cancer, 211–212 Benzydamine, 349 lymphoma, 306
mechanism of action, 127 BEP chemotherapy, ovarian solitary plasmacytoma, 315–316
ovarian cancer, 204 tumours, 222–223, 223b Bone marrow
side effects, 61, 127–128 Bereavement care, 73, 327 examination in leukaemia, 309,
Arrhythmias, cancer survivors, 63, Betel quid chewing, 77 309f, 312, 314
64t Bevacizumab, 41, 372, 373f failure, 309
Arsenic trioxide, 311 brain tumours, 270 response to chemotherapy, 33b,
Asbestos, 89, 107 breast cancer, 132 33f
Ascites, ovarian cancer, 198, colorectal cancer, 169 suppression, treatment-induced,
204 kidney cancer, 176 39b, 349
Astrocytoma, 270–271 lung cancer, 103 transplantation see stem cell
anaplastic, 266t, 270–271, malignant melanoma, 236 transplantation
273 mesothelioma, 110 Bone metastases
grade II (diffuse), 266t, 267f, toxicity, 370t breast cancer, 130–131
270–272 Bexarotene, 308 lung cancer, 106
pilocytic, 266t, 270–271 Bias, 366b lytic, 345–346, 346f
spinal cord, 280–281 Bicalutamide, prostate cancer, 187 malignant melanoma, 235
Ataxia, isolated, 329 Bile duct cancer, 152–154, 153f men with sclerotic, 288
Ataxia telangiectasia, 48t Biliary tract tumours, 150–154 prostate cancer, 183, 188f, 189
Atumumab, 308 Biological agents Bone pain
Atypical adenomatous hyperplasia head and neck cancer, 82 lung cancer, 91
(AAH), 90 lung cancer, 102–103 management, 69
Atypical ductal hyperplasia, 115 mesothelioma, 110 myeloma, 316
Atypical hyperplasia, oesophageal cancer, 141 Bone scan
endometrium, 205 as radiation modulators, 29 bone tumours, 246, 250, 251f
Atzpodien chemotherapy, 175 see also targeted systemic lung cancer, 92
Auer rods, 309, 309f therapy prostate cancer, 188f
Autonomic dysfunction, 329 Biopsy see tissue diagnosis Bone tumours, primary, 245–254
Axillary dissection, breast cancer, Bisphosphonates, 41 clinical features, 246, 246b
123–124, 128 breast cancer, 130–131 staging, 246, 247t
Axillary lymphadenopathy, hypercalcaemia of malignancy, Bortezomib
women with isolated, 287 340 myeloma, 318, 319t
Axillary nodes, 116–117, 117f myeloma, 319 oesophageal cancer, 141
prostate cancer, 189 Bowel Cancer Screening
Bladder cancer, 177–182 Programme (BCSP), 169
B investigations and staging, 177, Bowen disease, 239–241
Back pain, 328 179f, 179t management, 243
Baclofen, 70t, 71 management, 178–182, 178b, penis, 194
Bacterial infections, 334 180f vulva, 226
Bad news, breaking, 5–6, 6b secondary, 59t Bowenoid papulosis, 194
Barium enema, double contrast small cell, 182 Brachytherapy, 17, 18f
(DCBE), 161, 161f Bleomycin, 35 cervical cancer, 214, 217b
Barium studies, gastric cancer, lymphoma, 300b endometrial cancer, 207–211
142, 143f ovarian tumours, 223b head and neck cancer, 81
Barium swallow, 136, 136f testicular cancer, 192b penile cancer, 195–196
Barrett’s oesophagus, 134, 141 Blinding, 363–364 prostate cancer, 186, 187b
376
INDEX
Choroid plexus tumour, 274 heterogeneity of results, 367, endometrial cancer, 206
Choroidal metastasis, 333 367b gallbladder carcinoma, 151
Chronic health conditions, cancer patient recruitment, 357–358, gastric cancer, 143
survivors, 64 358b head and neck cancer, 79
Chronic lymphocytic leukaemia phases, 355–357, 359f kidney cancer, 174, 174f
(CLL), 312–314, 313t quality assessment, 368 lung cancer, 92, 94f
Chronic myeloid leukaemia stopping early, 363 lymphoma, 298, 302, 304f,
(CML), 311–312 CMV chemotherapy, 181, 181b 306f
Chronic myelomonocytic Co-morbidities, assessment of, 7 malignant melanoma, 232–233,
leukaemia, 314 Codeine, 68t 236
Cimetidine, 343 CODOX-M chemotherapy, 305, mesothelioma, 107–108, 108f
Cirrhosis of liver, 147–148, 150 305t oesophageal cancer, 136
Cisplatin, 35 Coeliac disease, 158 ovarian tumours, 198, 199f,
anal cancer, 171–172 Colectomy, prophylactic, 52–53, 221, 221f
bladder cancer, 181–182, 181b 52t pancreatic cancer, 155, 155f
brain tumours, 270b, 274–275 Colon cancer radiotherapy planning, 19–21,
cervical cancer, 215, 219 management, 53, 164f 21f
endometrial cancer, 211 resection margins, 12t small intestinal tumours, 159
gastric cancer, 146b, 147 see also colorectal cancer soft tissue sarcomas, 255
head and neck cancer, 81–82, Colonoscopy superior vena cava obstruction,
84b diagnostic, 161, 161f 337, 338f
lung cancer, 100–101, 101b, screening/surveillance, 52–53, testicular cancer, 190, 192f
104 169 thymoma, 111, 112f
mesothelioma, 109–110 Colony stimulating factors (CSFs), Concurrent controls, 361–362
oesophageal cancer, 138–140, 336, 337b Cone biopsy, 213, 292
140b Colorectal cancer (CRC), 159–170 Confidence intervals (CI), 365,
osteosarcoma, 247–249 aetiology, 160 366b
ovarian tumours, 223b epidemiology, 159–160 Conformal (3D) radiotherapy
skin cancer, 242 follow-up, 169 beam shaping, 22–23, 22b
testicular cancer, 192b inherited, 46t, 50–53, 51b, 160 planning, 21
thymoma, 113, 114b investigations, 161–162, Confusional state, acute, 332
Citalopram, 72 161f–162f Consent, informed
CK7, 288t management, 162–165, 164f, chemotherapy, 35–36, 36b
CK20, 288t 165t clinical trials participants,
Cladrabine, 314 pathogenesis and pathology, 357–358, 358b
Clark level, 233 160, 160f radiotherapy, 17
Clear cell carcinoma presentation, 160–161 Consolidation chemotherapy, 31b
endometrium, 205–206, 207t, prognosis and outcome, 169 Constipation, 39b, 69
212 screening, 169 Consultation, patient, 5–6, 6b
ovary, 197 secondary, 59t Continuous hyperfractionated
Clear-cell sarcoma, 255b staging, 162, 163f accelerated radiotherapy
Clinical approach, cancer patients, Complementary therapy, 40 (CHART), 26b, 28–29, 98
3–9 Complete response (CR), 44t Control groups, 361
Clinical Benefit, 42–43 Computed tomography (CT), 4, 4f Controls, concurrent or historical,
Clinical significance, clinical trial assessing response to treatment, 361–362
results, 365, 366b 43f Core biopsy, 10
Clinical target volume (CTV), bone tumours, 246 Coronary artery disease, cancer
20b brain metastases, 278, 279f survivors, 64t
Clinical trials, 355–359 brain tumours, 265, 266t, 276f Corticosteroids
advances, 358–359 cervical cancer, 213 adrenalectomized patients, 286
children and young adults, 326 cholangiocarcinoma, 153 brain tumours, 267–268
critical appraisal, 360–366, colonography, 161, 161f hypersensitivity reactions, 343
361b colorectal cancer, 161, 162f, leukaemia, 310
ethics, 357 169 myeloma, 319t
379
INDEX
External beam radiotherapy oesophageal cancer, 138–140, malignant potential, 262, 262b,
(EBRT), 15–16 140b 262t
External ear canal tumours, 85 pancreatic cancer, 156–157 management, 262–263, 262b
Extrapleural pneumonectomy penile cancer, 195 small intestine, 159, 262
(EPP), 108 rectal cancer, 167 stomach, 261f, 262
Extravasation, 344–345 Folinic acid (FA) Gastrointestinal system
Eye tumours, 85 breast cancer, 126b cancers, 134–172
malignant melanoma, 85, colorectal cancer, 163, 165t, late effects of treatment, 63, 63b
236–237 168–169 metastases in melanoma, 235
Follicular carcinoma of thyroid, Gefitinib, 370–372, 372f
282–283 lung cancer, 102–103
F Follicular lymphoma, 303–304, mesothelioma, 110
Faecal occult blood (FOB), 169 303t oesophageal cancer, 141
Familial adenomatous polyposis Follicular Lymphoma International toxicity, 370t
(FAP), 51–52, 51t, 158, Prognostic Index (FLIP Gemcitabine, 33–34
160 score), 302–303, 304b bladder cancer, 181, 181b
Familial cancer see inherited Follow-up combination therapy, 34b
cancer after treatment, 8–9 lung cancer, 101b
Fatigue, 39b, 72, 348 losses to, clinical trials, 364, mechanism of action, 34b, 34f
Febrile neutropenia see 365b ovarian cancer, 203
neutropenia, febrile during treatment, 7–8 pancreatic cancer, 156–157
Female genital system, cancers of, Fractionation, radiotherapy, 17, soft tissue sarcomas, 259b, 261
197–230 20b, 28–29 Gemtuzumab ozogamicin, 311
Fentanyl, 68t, 70f advances in, 28–29 Genetic counselling, 46–47, 47b
Fertility schedules, 25, 26b Genetic screening, 8, 45–46
children and young adults, see also continuous Genetic testing, predictive, 45–46
326 hyperfractionated Genetics, cancer, 45–57
female cancer survivors, 61b accelerated radiotherapy see also inherited cancer
preserving surgery, ovarian Fractures/impending fractures, Genitourinary system cancers,
tumours, 199–200, 221 345–346, 346f, 346t 173–196
see also infertility Fraudulent clinical research, 363, Germ cell tumours (GCTs)
Fibromas, ovarian, 224 364b central nervous system, 275
Fibromatosis, aggressive, 254 Frozen section, 10 children and young adults, 325
Fibrosarcoma, 259b Fungal infections, 334, 334f G-CSF indications, 337b
Fine needle aspiration (FNA), ovary, 220–223, 222f
10–11 testes, 189
Fitness for treatment, assessing,
G Germinoma, 269t, 275
6–7, 7t Gabapentin, 67–69, 70t Gestational trophoblastic disease
Flavopiridol, 314 Gallbladder carcinoma, 150–151, (GTD), 224–226, 225b,
Fludarabine 151b 225t
leukaemia, 313 Gallstones, 150, 152 Giant cell tumours of bone, 254
lymphoma, 304 Gamma-radiation therapy, 17 Gleason grading system, 183,
5-fluorouracil (5-FU), 33–34 Gastric cancer, 141–147 183b
administration, 37–38 investigations and staging, Gliadel wafers, 273
anal cancer, 171–172 142–143, 143b, 143f Glioblastoma multiforme (GBM),
bladder cancer, 181–182 management, 144–147, 145f, 266t, 270–271
breast cancer, 126b 146b biological agents, 270
carcinoid tumours, 158 resection margins, 12t, 144 recurrence and progression, 273,
colorectal cancer, 163, 165t, Gastric stump cancer, 147 274f
168–169 Gastrinoma, 158 treatment, 272–273
gallbladder cancer, 151 Gastrointestinal stromal tumours Gliomas, 270–274
gastric cancer, 146b, 147 (GIST), 159, 261–263 aetiology, 264
head and neck cancer, 81–82, assessing response to treatment, anaplastic (grade 3), 272–273
84b 42, 263 biological agents, 270
382
INDEX
Planning target volume (PTV), management principles, 290 Prostatectomy, radical, 185
20b, 23–25, 24b, 24f postpartum care, 292 Protocol, clinical trial
Plasmacytoma, solitary, 315–316, psychosocial support, 294 changes to original, 362–363
315f, 316b radiotherapy, 291, 291b pre-specified, 362
Platelet counts, 27–28, 336–337 surgery, 290–291 Psychological problems, cancer
Platelet transfusion, 336–337 systemic treatment, 291–292 survivors, 65
Platinum-based chemotherapy timing of delivery, 291 Psychosocial support see support
hypersensitivity reactions, in cancer survivors, 60–61, 60f, during treatment
342–343, 343t 61b PTEN mutations, 54–55
Merkel cell carcinoma, 243 termination, 290 Publication
ovarian tumours, 202–203, 223 Primary CNS lymphoma (PCNSL), duplicate, 366
see also carboplatin; cisplatin; 276–277, 306 foreign language, 367
oxaliplatin HIV-associated, 297, 306 Publication bias, 366, 366b, 368
Pleomorphic adenoma, parotid imaging, 266t, 306f Pulmonary function tests, 97,
gland, 86 Primary sclerosing cholangitis 100t, 299
Pleural effusions (PSC), 152–153 Pulmonary late effects of
lung cancer, 102, 108 Primitive neuroectodermal tumour treatment, 63
mesothelioma, 107–109, 108f (PNET), 245, 274–275
Plummer–Vinson syndrome, 135b, chemotherapy, 270b
141 staging, 246
Q
Pneumonectomy, 97, 100t Procarbazine, gliomas, 270b Question, clearly defined research,
extrapleural (EPP), 108 Proctitis, radiation, 350 362
Pneumonitis, radiation, 350 Proctocolectomy, prophylactic,
Poly (ADP-ribose) polymerase 13b, 52t
(PARP) inhibitors, ovarian Progesterone receptors (PgR)
R
cancer, 204 breast cancer, 115–117 R0, R1, R2 resections, 11
Poorly differentiated carcinoma, endometrial cancer, 205, 211 Radiation dermatitis, 348
with unknown primary, Progestogens, 72 Radiation dose
289 endometrial cancer, 211 bolus, 23
Positron emission tomography see also medroxyprogesterone foetal, 290–291, 291b
(PET) acetate; megestrol acetate prescription, 25, 27f
cervical cancer, 213 Prognostic factors, 5 tolerance, organs at risk (OAR),
colorectal cancer, 161–162 Progressive disease (PD), 44t 24, 25t
endometrial cancer, 206 Prophylactic cranial irradiation Radiation fibrosis, 350
gastric cancer, 143 (PCI), small cell lung Radiation recall, 348
head and neck cancer, 79 cancer, 104 Radiofrequency ablation (RFA)
Hodgkin’s disease, 299 Prostate cancer, 182–189 liver cancer, 150
lung cancer, 94–95, 97f aetiology, 183 liver metastases, 169
lymphoma, 308 hormone refractory (HRPC), malignant melanoma, 236
mesothelioma, 108 188 Radioimmunotherapy (RIT),
oesophageal cancer, 136 hormone treatment, 40–41, 41t, lymphoma, 308
ovarian tumours, 221 186–187 Radioiodine ablation
radiotherapy planning, 19–21, inherited, 46t, 183 side effects, 284b
21f investigations and staging, thyroid cancer, 283–284
Preclinical studies, 355 183–184, 184t Radionuclide treatment, 17
Prednisolone, myeloma, 319t management, 184–187 carcinoid tumours, 158
Prednisone, myeloma, 319t palliative treatment, 188–189 prostate cancer, 189
Pregabalin, 70t pathology, 183 Radiotherapy, 15–29, 16b
Pregnancy presentation, 183, 183b advances, 28–29
cancer during, 290–294 prognosis and survival, 189 beam shaping, 22–23, 22b,
care of baby, 292 Prostate specific antigen (PSA), 183 23f
diagnostic work-up, 290, cancer with unknown primary, bladder cancer, 180–181
291b 288 bone tumours, 249
future pregnancies, 292 predictive value, 183–184 brain metastases, 278
390
INDEX
brain tumours, 19, 268–270, second malignancies after, 28, resection margins, 12t, 167
269t, 272–275 58–59, 59t see also colorectal cancer
breast cancer, 121, 124–125, side effects, 17, 28 Recurrence
124b–125b, 129 management, 348–351 management, 8–9
central airway obstruction, 339 simulators, 19–21, 21f re-irradiation after, 29
cervical cancer, 214–218, 217b, skin cancer, 239, 240b, salvage surgery, 13
218f 241–244 Reed–Sternberg (RS) cells, 298
chemotherapy with see soft tissue sarcomas, 257–259, Refractory anaemia, 314
chemoradiation 261 Relationships, cancer survivors,
choroidal metastasis, 333 solitary plasmacytoma, 315, 65
colorectal cancer, 168 316b Relative risk (RR), 366b
curative, 15 spinal cord compression, Relative risk reduction (RRR),
delivery methods, 15–17 330–331, 330b, 331f 366b
endometrial cancer, 207–212, spinal cord tumours, 280–281, Renal cancer see kidney cancer
209b 281b Renal cell carcinoma (RCC),
errors in treatment set up, superior vena cava obstruction, 173–176
26–27 338 aetiology, 173
Ewing’s sarcoma, 252 supportive care, 27–28 hereditary papillary, 173
external beam (EBRT), 15–16 systemic, 42 management, 174–176
follow-up care, 28 testicular cancer, 190–191, pathology, 173–174
fractionation, 17, 20b, 25, 26b 192b, 192f prognosis and survival, 176
future directions, 29 thymoma, 112, 114b staging, 175t
gallbladder cancer, 151 thyroid cancer, 284–285 Renal pelvis, cancer of, 173–174,
gap correction, 28 treatment verification and 177, 182, 182t
gastric cancer, 146 corrections, 25–27, 26b, Research, cancer
gynaecomastia prophylaxis, 27f clinical trials, 355–359
187 vaginal cancer, 230 critical appraisal, 360–369
head and neck cancer, 19, volumes, 20b, 21f fraudulent, 363, 364b
80–81, 81b, 82t, 83b vulval cancer, 227–228 Research question, clearly defined,
indications, 15 see also brachytherapy; 362
informed consent, 17 chemoradiation Resection margins, 11, 12t
kidney cancer, 176 Radon, 89 Respiratory secretions, 73
late effects, 58–65 Randomization, 361–362 Respiratory symptoms, lung
lung cancer, 98, 100t, 101–102, allocation concealment, 364 cancer, 91
104 phase II studies, 356 Response Evaluation Criteria in
lymphoma, 300–301, 300b, Randomized controlled trials Solid Tumours (RECIST)
307–308 (RCTs) criteria, 42–43, 43b, 44t
malignant melanoma, 235, 237 critical appraisal, 360–366 Response to treatment, assessing,
mesothelioma, 108–109 design, 357, 357b 42–43, 43f
mode of action, 19b, 19f Rapirnase, mesothelioma, 110 RET proto-oncogene, 56, 285
oesophageal cancer, 137–138, Rasburicase, 342 Retinoblastoma, 45, 55, 326
139b, 141 RB1 gene mutations, 55 osteosarcoma, 245
osteosarcoma, 248 RECIST criteria see Response Retroperitoneal sarcomas, 260f,
ovarian tumours, 222 Evaluation Criteria in Solid 261
palliative, 15, 69 Tumours (RECIST) criteria Reviews
penile cancer, 195–196, 196b Reconstructive surgery, 13 narrative, 368
planning, 18–28, 22b, 22f, 24b, Rectal bleeding, cancer survivors, systematic, 366–369, 369b
24f 63 Rhabdomyosarcoma (RMS)
pregnancy, 291, 291b Rectal cancer alveolar, 255b, 325
prostate cancer, 185–189, 185b, investigations and staging, 161, children and young adults, 325
186f 161f–162f embryonal, 325, 325f
rectal cancer, 167, 168t management, 165–168, 166f, late effects of treatment, 58–59
repeat, 29 168t management, 258–259
sarcomas, 253b presentation, 160–161 Richter’s syndrome, 313
391
INDEX
Risk of malignancy index (RMI), Sertoli–Leydig cell tumours, Sodium valproate, 70t
ovarian tumours, 198, 198b ovarian, 224 Soft tissue sarcomas (STS),
Risk reduction surgery, 13, 13b Sex cord stromal tumours, 254–263
Rituximab, 41 223–224 aetiology, 254
hypersensitivity reactions, 343t Sezary syndrome (SS), 307–308 children and young adults, 325
leukaemia, 311 Shields, radiation, 23 diagnosis, 255
lymphoma, 300, 304–306 Sigmoidoscopy, 161 management, 253b, 255–260,
Roach formula, 183, 183b Signs of cancer, warning, 3, 3b 257f
Robotic assisted surgery, 14 Sister Mary Joseph nodule, 142 pathology, 255, 255b
Rodent ulcer, 237 Skin, radiation reactions, 348 presenting features, 255
Roswell Park, modified Skin cancers, 231–244 prognostic factors, 258, 259b
chemotherapy, 165t adnexal carcinomas, 244 specific subtypes, 260–263
basal cell carcinoma, 237–239 staging, 255, 256t
eyelids, 85 Solitary plasmacytoma, 315–316,
S lymphoma, 244, 307–308, 307f 315f, 316b
Salivary gland tumours, 86 malignant melanoma, 231–236 Somatostatin analogues, thymoma,
Salpingo-oophorectomy, bilateral Merkel cell carcinoma, 243–244 113
(BSO), 207, 224 squamous cell carcinoma, 12t, Sorafenib, 372–373, 373f
Salvage surgery, 13 239–243, 241f kidney cancer, 176
Sample size Skin metastases, malignant liver cancer, 150
adequacy, 365–366 melanoma, 235–236 malignant melanoma, 236
slippages, 364, 365b Small and non-small cell lung mesothelioma, 110
Sanctuary sites, chemotherapy, cancer, mixed, 105–106 Sperm banking, 60
35b Small cell carcinoma Spinal cord compression,
Sarcomas, 245–263 bladder, 182 metastatic (MSCC),
head and neck, 88 oesophagus, 141 328–332
orbital, 85 Small cell lung carcinoma (SCLC), intramedullary, 328, 332
paediatric, teenage and young 90 investigations, 329, 329f
adult, 325 management, 101b, 103–105, management, 329–331
radiotherapy, 253b 103f paediatric, 332
secondary, 58, 59t palliative care, 106–107 prognosis, 331–332
small intestine, 159 paraneoplastic syndromes, recurrence, 332
Screening 91–92 Spinal cord tumours, 280–281,
cancer, 8 prognostic factors and survival, 280b–281b
genetic, 8 105 Spindle cell carcinoma, skin,
Sebaceous carcinoma, 244 staging, 95b 241–242
Second cancers, 28, 58–59, 59b, Small intestine Splenectomy, 314
59t, 60f lymphoma, 158–159 Sputum cytology, 92
brain, 264 malignant tumours, 158–159 Squamous cell carcinoma (SCC)
Hodgkin’s disease, 59, 301–302 metastases, 159 anal canal, 170
Seminomas, 189, 194 obstruction, ovarian cancer, cervix, 213
investigations, 189–190 204 cutaneous, 12t, 239–243,
postoperative management, Small molecule drugs, 41–42 241f
190–194, 191f, 192b Smoking head and neck, 77
Sensitivity analysis, 368 gastrointestinal cancers and, aetiology, 77
Sensory deficits, 328 152, 154, 170 management, 81–82
Sentinel node biopsy (SNB), 11, head and neck cancer and, 77 pathogenesis and pathology,
12f lung cancer and, 89 77–78
breast cancer, 123–124 renal cancer and, 173 see also head and neck cancer
malignant melanoma, 233 Smooth muscle spasm, 70t kidney, 174
Serous cystic neoplasms, pancreas, Social issues, cancer survivors, 65 lip, 243
158 Socioeconomic status, head and lung, 90
Serous papillary carcinoma, neck cancer, 77 middle ear, 85
uterine (UPSC), 205–206, Sodium 2-mercaptoethane nose and paranasal sinuses, 85
392 207t, 212 (mesna), 346–347 oesophagus, 134
INDEX
head and neck cancer, 79 Vesicant drugs, 344, 345t Whole brain radiotherapy
lung cancer, 95, 98f Vestibular schwannoma (WBRT), brain metastases,
ovarian cancer, 198 (acoustic neuroma), 269t, 278–279
pancreatic cancer, 155 275 Wide local excision (WLE)
small intestinal tumours, 159 VHL gene, 55 basal cell carcinoma, 239
Ultraviolet (UV) radiation Video-assisted thoracoscopy breast cancer, 123–124
head and neck cancer risk, 77 (VATS), 96, 111 Wilms’ tumour, 323, 324f
skin cancer risks, 231, 237, 239 Vinblastine World Health Organization
Ureter, carcinoma of, 177, 182, bladder cancer, 181, 181b (WHO)
182t lymphoma, 300b analgesic ladder, 66, 67f
Urinary diversion, 180 ovarian tumours, 223b performance score, 6–7, 7t
Uterine cancer, secondary, 59t Vinca alkaloids, 32b, 34
Uterine sarcomas, 205, 260–261 Vincristine, 34
leiomyosarcomas (LMS), brain tumours, 270b, 274–
X
260–261 275 X-rays
undifferentiated, 261 Ewing’s sarcoma, 251 external beam radiotherapy,
Uterine serous papillary carcinoma leukaemia, 310 15–16
(UPSC), 205–206, 207t, myeloma, 319t impending fractures, 346,
212 Vinorelbine, lung cancer, 100– 346f
101, 101b myeloma, 316, 317f
Viral infections, head and neck primary bone tumours, 246,
V cancer risk, 77 248f, 250, 251f, 253f
VAC chemotherapy, Ewing’s Virchow’s node, enlarged, 142 see also chest X-ray
sarcoma, 251 Visual loss, acute, 333 Xerostomia, 349
Vaginal cancer, 228–230, 229b Vomiting see nausea and
Vaginal vault, endometrial cancer vomiting
recurrence, 212 Von Hippel–Lindau syndrome
Y
Vascular endothelial growth factor (VHL), 55, 56t, 173 Yolk sac tumours (YST), 220
(VEGF), 372, 373f Vote counting, 368 Young adults
Vasoactive intestinal polypeptide Vulval cancer, 226–228, 227b cancers see teenage and young
tumour (VIPoma), 158 Vulvar intraepithelial neoplasia adult (TYA) cancers
Vatalanib, mesothelioma, 110 (VIN), 226 poorly differentiated carcinoma
VelP chemotherapy, ovarian with midline distribution,
tumours, 223b 289
Venlafaxine, 72
W
Venous thromboembolism, Warning signs of cancer, 3, 3b
ovarian cancer, 201b Watchful waiting, prostate cancer,
Z
Verrucous carcinoma 185 Zolendronic acid, 61–62
penis, 194 Wedges, radiotherapy, 23 hypercalcaemia of malignancy,
skin, 241 Weight loss, 5, 348–349 340
vulva, 226 Whipple procedures, 156 prostate cancer, 189
395