A Systematic Review On The Culture Methods and Applications of 3D Tumoroids For Cancer Research and Personalized Medicine
A Systematic Review On The Culture Methods and Applications of 3D Tumoroids For Cancer Research and Personalized Medicine
https://doi.org/10.1007/s13402-024-00960-8
REVIEW
Abstract
Cancer is a highly heterogeneous disease, and thus treatment responses vary greatly between patients. To improve therapy
efficacy and outcome for cancer patients, more representative and patient-specific preclinical models are needed. Organ-
oids and tumoroids are 3D cell culture models that typically retain the genetic and epigenetic characteristics, as well as
the morphology, of their tissue of origin. Thus, they can be used to understand the underlying mechanisms of cancer
initiation, progression, and metastasis in a more physiological setting. Additionally, co-culture methods of tumoroids and
cancer-associated cells can help to understand the interplay between a tumor and its tumor microenvironment. In recent
years, tumoroids have already helped to refine treatments and to identify new targets for cancer therapy. Advanced cultur-
ing systems such as chip-based fluidic devices and bioprinting methods in combination with tumoroids have been used
for high-throughput applications for personalized medicine. Even though organoid and tumoroid models are complex in
vitro systems, validation of results in vivo is still the common practice. Here, we describe how both animal- and human-
derived tumoroids have helped to identify novel vulnerabilities for cancer treatment in recent years, and how they are
currently used for precision medicine.
Keywords Cancer · 3D models · Preclinical models · Organoids · Tumoroids · Precision medicine · Co-culture ·
Bioprinting · Fluidic devices
                                                                                                                  13
                                                                                                                     J. Kalla et al.
1.1 Model systems for research                                    Although a lot of research has been done on 3D models
                                                                  derived from stem cells in the past [15], the group of Hans
One of the biggest challenges in science is the representative    Clevers pioneered the successful establishment of organoids
modeling of multicellular tissues, dynamic interactions, and      in the modern era [14]. These organoids were generated in
the complex mechanisms underlying disease. Therefore,             2009 from murine intestinal stem cells, and since then many
scientists mostly rely on different model systems ranging         different research groups have adapted the establishment
from cell lines to animal models, to representative models        protocol to generate an extensive collection of organoid
derived from primary tissue [1].                                  models from both animals and humans [16].
   2D cell lines have been established from a diverse range          Typically, primary surgical resection material is cut into
of malignant diseases but also from a selection of cell types     small fragments using scalpels, followed by an enzymatic
in the healthy human body and are a widely used model             digestion to dissociate the tissue into single cells or small
system. Because of easy handling for in vitro cell culture        cell clumps [17, 18]. However, as access to tissue samples
and simple introduction of genetic modifications, they can        can be anatomically and logistically limited, stem cells
deliver meaningful insights on specific research questions        derived from fine needle aspirates of tissues [19–21], fluid
[2]. However, these cell lines consist of only one cell type      samples such as urine [22], ascites [23], broncho-alveolar
optimized for long-term culture. Additionally, as the culti-      lavage fluid [24], and bile [25], or enriched circulating
vation in a monolayer can alter the shape, polarization, and      tumor cells (CTCs) [26] might provide a minimally invasive
global signalling of cells, 2D cell lines do not adequately       alternative for organoid generation.
reflect the physiological situation in organs [3, 4]. There-         Organoids are mostly cultivated in 3D ECM hydrogel
fore, research relying solely on cell lines does not success-     domes such as basement membrane extract, Matrigel® [27],
fully translate from bench to bedside [5, 6].                     or Geltrex®, in addition to a variety of xeno-free synthetic
   Animal models, which better reflect the cellular complex-      hydrogels [28]. Organoid culture medium provides a vari-
ity and molecular crosstalk of organs in the human body, are      ety of niche-specific growth factors matched to the tissue
frequently used to study cells in their physiological context.    of origin to ensure optimal growth conditions [17, 29, 30].
However, animals and humans are still inherently different        Common key components include activators of the canoni-
so that not all biological processes can be modeled accu-         cal WNT pathway (R-Spondin), MAPK pathway (EGF),
rately. Moreover, animal models are more complicated and          and inhibitors of TGF-β (A83-01) as well as BMP signaling
time consuming to work with than in vitro cell lines, in addi-    (Noggin) to allow stem cell renewal and proliferation [11,
tion to the ethical issues connected to the use of animals for    14, 17].
research [7].                                                        Stable organoid lines can be expanded long-term (> 10
                                                                  passages) in vitro, and cryopreserved [31]. Of note, even
1.2 Organoids                                                     though conventional submerged organoid cultures exclu-
                                                                  sively enrich for epithelial cells and fail to retain stromal
Organoids are one model system that combines several              components [32], organoids do recapitulate the tissue archi-
advantages of in vitro 2D cell culture and in vivo animal         tecture on a microscale, and can mimic the function of the
models [8]. Even though commonly used 3D spheroids are a          organ they resemble, while stably maintaining mutational
promising tool for cancer research, they are cell line-derived    and epigenetic states in line with cellular signaling [33–35].
aggregates that do not self-organize or differentiate into
multiple cell types [9]. In contrast, organoids are 3D self-      1.2.2 Organoids for cancer research: tumoroids
organizing cell clusters that can be derived from embryonic
stem cells, tissue-specific adult stem cells, or from induced     Organoids are used in many different research areas, includ-
pluripotent stem cells (iPSCs) from both animals and              ing reconstructive medicine, evolutionary biology, and
humans. These stem cells, which are commonly embedded             pathogen-host interactions, summarized in previously pub-
in an extracellular matrix (ECM), can then further differen-      lished reviews [36–41]. In recent years, tumoroids derived
tiate into different types of epithelial cells specific for the   from both animals and humans have been intensively used
tissue of interest [10–14]. In addition to stem cell-derived      for preclinical cancer modeling, with a focus on cancers
organoids, tumor-derived organoids, so called tumoroids,          originating from epithelial cells (Fig. 1). However, efforts
have been successfully generated for various cancer entities      have been made to also establish tumoroids from non-epi-
and provide valuable tools for cancer research.                   thelial cancers and rare cancer types, for example melanoma
13
A systematic review on the culture methods and applications of 3D tumoroids for cancer research and…
Fig. 1 3D model systems in cancer research. Applications of both         Comparison of different 3D model systems, including organoids, co-
human- and animal-derived tumoroids in cancer research (top).            culture models, microfluidic devices, and in vivo models, highlighting
Tumoroids can be used for either cancer modeling with a focus on the     the advantages and disadvantages of each model (bottom). Created
tumor microenvironment (TME), or as a tool for personalized medi-        with BioRender.com
cine and drug discovery based on the mutational landscape of patients.
[42], glioblastoma [43, 44], rhabdomyosarcoma [45], epi-                 organoids and tumoroids could be used for both answering
thelioid sarcoma [46], and rhabdoid cancer [47].                         basic questions of tumorigenesis and developing a personal-
   Disease modeling using tumoroids allows for the inves-                ized medicine approach to help advance the field of cancer
tigation of different hurdles of oncology such as tumor ini-             research.
tiation and driver events of cancer, tumor heterogeneity or                 Given recent advancements in organoid technologies, we
development of efficient therapies [10, 48–50]. By estab-                here summarize the latest developments in the field, out-
lishing healthy organoids and altering cancer type specific              lining various methods and practical applications of these
genes in vitro, the transformation from healthy to cancerous             innovative model systems. We will focus on current uses for
cells can be studied in a 3D model focusing on different                 cancer research including both human- and animal-based
tissues and diseases. Additionally, by generating organoids              methods. Furthermore, we will highlight recent work that
and tumoroids directly from both healthy and tumor tissue,               has improved preclinical cancer models by using tumoroids
cancer driver mutations can be identified and used to dis-               in combination with other cell types to model the tumor
cover patient-specific therapeutic targets [33, 51]. This way,           microenvironment (TME). Moreover, studies focusing on
                                                                                                                                   13
                                                                                                                         J. Kalla et al.
advanced 3D culture systems based on fluidic devices and            models have been established and used across all fields of
bioprinting, and grafting tumoroid models into animals for          cancer research (Table 1). Importantly, the availability of
in vivo based studies, will be summarized. Finally, existing        normal tissue samples and thus healthy organoid lines from
limitations are discussed, and future developments of the           animal models allows for the evaluation of adverse side
field are highlighted.                                              effects of cancer therapies on healthy tissue [52].
                                                                       In the past, cancer research mainly relied on genetically
                                                                    modified mouse models (GEMMs) targeting genes com-
2 In vitro culture methods and applications                         monly mutated in human cancers [93]. Thus, a variety of
for cancer research                                                 organoids and tumoroids from these mouse models exist,
                                                                    which can further be easily genetically modified in vitro.
2.1 Conventional ECM-drop culture                                   Two recent studies focused on the development of preclini-
                                                                    cal 3D models for ovarian cancer based on murine cancer
2.1.1 Animal-derived tumoroids in cancer research and               models [94, 95]. The knock-out of ovarian cancer-specific
their application                                                   genes was induced using either a Cre recombinase-based
                                                                    system or CRISPR/Cas9 in organoids derived from healthy
As tissue samples from animals often are easier accessible          murine tissue in vitro, leading to their malignant transfor-
than most human biopsies, and as a big variety of murine            mation. The group of Zhang et al. hereby focused on the
cancer models exist, animal-derived tumoroids can be espe-          influence of the mutational heterogeneity on treatment
cially useful when human-derived tumoroids are not avail-           response, and showed that both tumoroids and mice with
able. Therefore, many murine and other animal-derived 3D            the same mutational background can be treated success-
                                                                    fully with identical combination therapies [94]. The group
Table 1 Summary of animal-derived tumoroid models, including        of Löhmussaar et al. established a tumor progression model
murine, canine, and porcine 3D models for the most commonly diag-   for high-grade serous ovarian carcinoma from two different
nosed cancer types
                                                                    epithelial lineages of mouse ovaries. Their findings support
Species        Cancer Type               Year         References
Mouse          Breast Cancer             2023         [53, 54]
                                                                    the dual-origin hypothesis of these tumors and confirmed
                                         2022         [55]          lineage-dependent differences in drug sensitivities [95].
                                         2020         [56]          Thus, these studies highlight how murine tumoroids can be
                                         2016         [57]          used as a model for identifying novel genotype-informed
               Lung Cancer               2021         [58]          treatments, and for investigating the differences and vulner-
                                         2020         [59, 60]
                                         2018         [61]          abilities of tumors arising from distinct cell types, leading to
                                         2017         [62]          more patient-specific and efficient therapy options.
               Colorectal Cancer         2022         [63–65]          Another approach using tumoroids derived from a pros-
                                         2020         [66, 67]      tate cancer (PCa)-specific mouse model was reported by
               Prostate Cancer           2022         [68]          Chan et al., who investigated genetic driver mutations of
                                         2019         [69]
                                         2018         [70]
                                                                    treatment-induced transformation and plasticity [68]. The
                                         2016         [71]          group induced malignant transformation of healthy organ-
                                         2015         [72]          oids by knocking out the PCa-specific genes Pten, Rb1, and
                                         2014         [73, 74]      Tp53 in vitro and showed that over time the resulting tumor-
               Gastric Cancer            2022         [75]          oids acquired an intermediate luminal-basal phenotype. An
                                         2021         [76]
                                         2019         [34, 77]      epithelial to mesenchymal (EMT) signature was further sup-
                                         2014         [78]          porting tumoroid plasticity, a phenomenon that has recently
               Liver Cancer              2021         [79]          also been investigated in pancreatic cancer (PaCa) organ-
                                         2020         [80]          oids [96]. Of note, the authors reported that the PCa tumor-
                                         2019         [81, 82]
                                                                    oid plasticity was further induced by androgen ablation (the
Dog            Breast Cancer             2022         [83]
                                         2017         [84]          suppression or blockage of the androgen pathway using
               Lung Cancer               2023         [85]          chemical compounds), the most common treatment for PCa,
               Colorectal Cancer         2019         [86]          and identified JAK/STAT and FGFR signaling as the main
               Prostate Cancer           2017         [87]          drivers [68]. Since the inhibition of these two pathways con-
               Bladder Cancer            2020         [88]          verted the tumoroids back to an androgen ablation-sensitive
                                         2019         [89]          more luminal phenotype, PCa patients may benefit from the
               Thyroid Cancer            2023         [90]          same treatment. The researchers also tried to confirm their
                                         2021         [91]
                                                                    results in human PCa tumoroids. However, it is difficult to
Pig            Intestinal Cancer         2017         [92]
13
A systematic review on the culture methods and applications of 3D tumoroids for cancer research and…
maintain human prostate organoids and tumoroids in long-                 3D models were then used to show that trametinib, a drug
term cultures, and the results did not overlap [68, 97, 98].             mainly used for melanoma treatment [103], and extracts
More studies are needed to verify these findings, but PCa is             from the Chaga mushroom [104], could be a potential
a good example for how animal-derived 3D models could                    therapy option for patients with BlCa. Recently, the same
be used as an alternative model for human cancer.                        researchers also established canine healthy lung and lung
    To investigate metastasis formation in addition to tumor             cancer 3D models that resemble human disease as a new
heterogeneity, two of the main factors for poor overall sur-             model for molecular analysis and drug testing of lung can-
vival of breast cancer (BrCa) patients, tumoroids from the               cer [85]. It can be expected that additional organoid systems
C3(1)-TAg BrCa mouse model were used [54]. Individual                    will be established from non-conventional animals in the
tumoroid lines established from the same primary tumor                   near future, some of which might have high translational
showed heterogeneous invasive mechanisms that were clas-                 impact for human cancer [105, 106].
sified into collective invasion or dissemination of single
cells. Interestingly, KRAS expression was required for                   2.1.2 Human-derived tumoroids in cancer research and
both mechanisms, and ERK inhibition blocked both inva-                   their application
sive processes [54]. Additionally, it was shown that col-
lective migration of BrCa cells is mediated by a keratin 14              Patient-derived tumoroids (PDTs) maintain the characteris-
and cadherin 3-positive subpopulation of leader cells [53].              tics of the primary tumor and can thus be used as a model
These cells have an enhanced protrusive activity and inter-              to identify the most efficient cancer treatment for individual
act with the TME to initiate invasion, and could become                  patients, laying the foundation for personalized medicine
a novel target for preventing or treating BrCa progression.              [107]. Therefore, tremendous efforts have been made to
However, murine 3D cell models can not only be used to                   generate organoids and tumoroids from diverse tissues and
test the effectiveness of drugs, but also of other anti-cancer           tumor types. Until now, PDTs from more than 20 different
treatments like radiation. After irradiating murine intestinal           carcinoma types, including BrCa, lung cancer, and colorec-
organoid models, Du et al. demonstrated that the induction               tal cancer (CRC) have been established (Table 2).
of inflammation with Zymosan-A promoted the regenera-                        Due to their advantages over 2D cell culture, PDTs are
tion of intestinal stem cells by up-regulation of ASCL2 [52].            widely used for drug screening and therapy response pre-
Thus, Zymosan-A may be an effective radio-protective drug                diction of patients [17]. Additionally, as it was shown that
for the prevention of harmful side effects on surrounding                both animal- and human-derived tumoroids do not acquire
healthy tissue and treatment of ionizing-radiation-induced               additional mutations during long-term culture in vitro [148],
intestinal injury.                                                       numerous studies have already proven the usefulness of
    In general, the results obtained using murine organoid               such 3D systems for precision medicine including CRC
systems that mimic human disease adequately reflect human                [113, 149–151], PaCa [152–154], as well as rare cancer
physiology and mechanisms [52–54, 68, 94, 95]. However,                  types such as Merkel cell carcinoma [147]. In CRC PDTs,
rodents and mammals are inherently different, which can                  the response of patients to oxaliplatin was predicted with
lead to unexpected discrepancies during translation to the               a sensitivity of 70% and a specificity of 71% [155]. Addi-
clinic [99, 100]. Recently, the use of organoids and tumor-              tionally, transcriptome analysis revealed that differences
oids derived from farm or companion animals has become                   in oxaliplatin response were mediated by distinct genetic
more popular in the field of cancer research, as animals like            features, and 18 specific genetic alterations were identified
cattle, horses, pigs, monkeys, dogs, and cats have a more                as a potential biomarker panel for oxaliplatin resistance
similar anatomy to humans [101]. One big difference to                   in CRC patients to aid clinical decision making. Another
rodents is that these animals can develop spontaneous can-               study focusing on the resistance of PaCa patients to neo-
cer lesions, which makes them more suitable to study the                 adjuvant chemotherapy confirmed a high heterogeneity in
mechanisms of cancer initiation and metastasis [83]. For                 patient responses, supporting the notion that personalized
example, in dogs bladder cancer (BlCa) arises spontane-                  medicine approaches might be more efficient [156]. Hennig
ously with similar pathology and genetics to human disease               et al. hypothesized that the observed resistance is mediated
[89], while murine tumoroids do not fully reflect the char-              by resistant clones residing in the tumor that get enriched
acteristics of human BlCa [102]. Therefore, Elbadawy et                  under systemic treatment. In addition to intrinsic resistance
al. focused on canine tumoroids derived from CTCs from                   of tumors, acquired resistance can also hinder efficient treat-
urine samples to generate a more representative model to                 ment. Most research on the underlying mechanisms of this
study BlCa [89]. Later the same group also established                   phenomenon have been generated using 2D cell lines, but
canine healthy bladder organoids to study the transforma-                recently resistant tumoroid models have been established as
tion of healthy cells to cancer cells [102]. The canine BlCa             a more representative system [157–159].
                                                                                                                             13
                                                                                                                     J. Kalla et al.
Table 2 Summary of human tumoroid models for the most commonly   of ovarian cancer PDTs derived from different sites of the
diagnosed cancer types
                                                                 same primary tumor for a small patient cohort [162]. The
Cancer type                             Year       Reference
                                                                 group reported that individual tumoroids showed a differen-
Breast cancer                           2022        [108]
                                        2020        [109]        tial response of 31%, defined as a more than 10-fold change
                                        2018        [110]        in IC50 value, to the same treatment, proving inter-patient
Lung cancer                             2019        [24, 111]    heterogeneity. Additionally, individual tumoroids from
                                        2017        [112]        the same primary tumor showed high differences in drug
Colorectal cancer                       2022        [113]        response in six out of seven cases, suggesting that PDTs
                                        2016        [114]
                                        2011        [11]         genetically maintain the heterogeneity and drug sensitiv-
Prostate cancer                         2022        [97]         ity of the original tumor [162]. Interestingly, it was recently
                                        2021        [115]        shown that a more physiological culture medium changes
                                        2014        [26]         the drug response of ovarian tumoroids compared to com-
Gastric cancer                          2018        [116–118]    monly used organoid medium, highlighting the necessity for
Liver cancer                            2018        [19]
                                                                 careful selection of experimental conditions [166]. Using a
                                        2017        [119]
Cervical cancer                         2022        [120]
                                                                 similar approach, the intra-tumoral drug response differ-
                                        2021        [121]        ences of liver cancer were investigated [163]. In this study,
Esophageal cancer                       2021        [122]        27 PDTs were generated from different areas of five primary
                                        2019        [123]        lesions and a total of 129 cancer drugs were tested. Even
                                        2018        [124]        though a high inter-patient and intra-tumoral heterogeneity
Thyroid cancer                          2023        [125]
                                                                 was confirmed, the researchers identified several pan-effec-
                                        2022        [126]
                                        2021        [127]        tive drugs that showed an inhibitory effect for most of the
Bladder cancer                          2023        [128]        tested tumoroids [163].
                                        2018        [129]           The high heterogeneity of cancer is a significant prob-
Pancreatic cancer                       2018        [130]        lem for efficient treatment; however, metastasis formation
                                        2017        [112]        is still the main reason for cancer related death. To eluci-
                                        2015        [131, 132]
                                                                 date the differential drug responses between primary tumors
Endometrial cancer                      2023        [133]
                                        2019        [134]        and matched liver metastases of CRC, Mo et al. generated
                                        2017        [112]        tumoroids from both samples of 25 patients, and showed
Ovarian cancer                          2020        [135, 136]   that the intra- and inter-patient heterogeneity is reflected by
                                        2019        [23]         the in vitro 3D models [167]. Interestingly, although tran-
Glioblastoma                            2020        [137]
                                                                 scriptomic analysis revealed differences between primary
                                        2019        [138]
                                        2016        [139]        and metastatic tumoroids, drug sensitivities were highly
Head and neck squamous cell carcinoma   2023        [140]        consistent. Thus, the authors suggested that the response of
                                        2019        [141]        metastatic lesions to specific drugs could be predicted using
                                        2018        [142]        tumoroids derived from primary tissue for a personalized
Mesothelioma                            2018        [143]        medicine approach [167].
Renal cancer                            2022        [144]
                                                                    The predictive potential of PDTs and their translation to
                                        2021        [145]
                                        2019        [146]        the clinics has also been investigated for various other can-
Merkel cell carcinoma                   2022        [147]        cer types, such as PaCa [168], and brain cancer [169], two
                                                                 highly aggressive cancer types with limited or inefficient
   As confirmed by the recent improvements of transcrip-         treatment options. In a translational approach, tumoroids
tome and proteome analysis on single cell level, one of the      of different stages of PaCa and brain tumors were used to
remaining problems for efficient cancer treatment is the         predict the optimal treatment for each patient in a clinically
intra-tumoral and inter-patient heterogeneity [160, 161].        relevant time frame [168, 169]. Even though both studies
Studies showed that between different regions of the same        confirmed that tumoroids reflect inter-patient heterogeneity
tumor, drugs can have varying inhibitory effects [162, 163].     and patient-specific drug responses, limitations of varying
This highlights the fact that biopsies might not be repre-       efficiencies of PDT generation, time management, but also
sentative for the drug response of the whole tumor, and          cost, have to be overcome for direct translation of tumoroid
that specific combinatorial therapies might be necessary to      research to the clinics. Especially for rare cancers, where
target all tumor subclones [164, 165]. To analyze whether        a major problem for efficient treatment is the lack of prog-
PDTs can reflect the intra-tumoral heterogeneity of a pri-       nostic and diagnostic biomarkers due to small patient num-
mary tumor, De Witte et al. compared the drug responses          bers [170, 171], tumoroids might be suitable to improve
13
A systematic review on the culture methods and applications of 3D tumoroids for cancer research and…
treatment response and survival rates of patients [172]. For             2.2 Co-cultures and advanced culturing systems
example, PDTs of PD-L1 negative mucinous adenocarci-
noma of the appendix were used to predict response to che-               Recent advances in the field of cancer research highlight the
motherapeutic drugs and targeted therapy for one individual              critical importance of not only the tumor itself but also its
patient [173]. PDT drug response correlated well with the                surrounding TME. The interplay between the tumor and the
patient response, and the tyrosine kinase inhibitor dasatinib            TME is a key determinant in tumor growth and progres-
was identified as a possible treatment option for this patient.          sion, making it an essential focus for comprehensive stud-
   In addition to evaluating the efficiency of commonly used             ies [180–182]. Traditional 2D or 3D cell culture models are
treatments for patient-specific precision medicine, PDTs can             limited in their ability to fully capture these complex inter-
also be used to test the efficacy of novel treatment regimes.            actions, prompting the development of more sophisticated
As an example, lung cancer PDTs are highly sensitive to the              methods. In recent years, three innovative approaches have
tyrosine kinase inhibitors dabrafenib and trametinib, which              emerged to model the TME in a more physiologically rel-
are commonly used for melanoma treatment, suggesting                     evant manner. This chapter will discuss: (1) the integration
these drugs as a novel therapy for lung cancer [174].                    of co-culture techniques with an air-liquid interface (ALI),
   In summary, several studies have provided clinically rel-             which allows for relevant interactions between tumor cells
evant evidence that PDTs can be a suitable model for a pre-              and their environment; (2) the application of bioprinting to
cision medicine approach to overcome the problem of high                 create tumoroids including cells of the TME, which offer
heterogeneity in drug responses between patients. In line                a more realistic, 3D structure for studying tumor behavior;
with this, PDTs have also been used to identify pre-existing             3) the incorporation of microfluidic channels, which can be
and acquired resistance to commonly used drugs, to avoid                 used independently or combined with bioprinting, to simu-
treatment of patients with inefficient therapies.                        late dynamic factors like blood flow or targeted drug admin-
   PDTs are not only suitable to predict the response of                 istration (Fig. 2A).
tumor cells to chemotherapy or targeted drugs, but also to
other treatments like radiotherapy [141, 175–178]. Radia-                2.2.1 Co-cultures
tion directly leads to the induction of single-strand and
double-strand breaks in DNA, as well as the generation of                To investigate the interplay between cancer cells and their
reactive oxygen species and the upregulation of oxidative                surrounding environment, advanced co-culture models,
stress signaling pathways, and is thus commonly used as                  including the culturing of tumoroids with cancer-associated
a combinatorial treatment with chemotherapy [179]. For                   fibroblasts (CAFs) [187, 188], lymphocytes [189], and
patients with locally advanced rectal cancer, the standard               myeloid cells such as macrophages [190–192], or even bac-
treatment is neoadjuvant chemoradiation followed by total                teria [193] have been used. Indirect cell interactions can be
mesorectal excision of the tumor [175]. Drug response of                 mimicked by the addition of cell-conditioned media, which
rectal PDTs matched chemoradiation responses in patients                 contains secreted molecules, to another cell type. However,
with an accuracy of up to 84% [175, 176], and a sensitiv-                this co-culture system only allows for single-directional
ity of 78.01% with 91.97% specificity [176]. The predictive              response, which can be overcome by the use of transwell
potential of PDTs for radiotherapy has also been confirmed               cultures. Individual cell types are separated via a mesh
for head and neck squamous cell carcinoma (HNSCC),                       leading to diffusion of secreted molecules and therefore
where the combination of the EGFR inhibitor cetuximab                    to a multi-directional interaction [13, 188]. However, as
and radiotherapy resulted in increased cell death in vitro,              direct cell-cell contact is essential for various physiological
and showed higher efficacy compared to radiotherapy alone                processes, indirect co-culture systems can only be used to
[141]. A recent study from the same researchers confirmed                investigate the effect of metabolites, cytokines, and other
the predictive potential of HNSCC tumoroids by correlat-                 secreted molecules. In direct co-culture systems different
ing tumoroid therapy response with patient clinical response             cell types are embedded in the same ECM and then either
to model the efficacy of chemoradiation for these patients,              submerged in culture medium or maintained as an ALI.
and the further use for biomarker selection and validation               Because of the direct cell-cell contact, different aspects
[177]. In addition, PDTs offer an advanced model to study                of the interaction between the tumor and its TME can be
the dynamics and mechanisms of radioresistance. Glio-                    studied. However, as it is more complex to isolate specific
blastoma PDTs reflected in vivo resistance mechanisms                    cell types for downstream analysis, single cell read outs or
mediated by treatment-induced senescence to combination                  optical techniques are required to efficiently analyze cell-
treatment with temozolomide and radiation, suggesting the                cell dependencies or their crosstalk [187, 190, 194, 195].
use of PDTs for studying the underlying mechanisms of                    As important factors normally found in the TME are lost
drug resistance [178].                                                   upon dissociation of the tumor tissue and the subsequent
                                                                                                                             13
                                                                                                                                 J. Kalla et al.
Fig. 2 Co-culture techniques for tumoroid models. (A) Overview          of sections of bioprinted tumoroids stained for CD31 (yellow), VIM
of state-of-the-art co-culture techniques in cancer research. (B) ALI   (red) and KRT8/18 (green) (bottom) [185]. (D) Schematic (middle)
tumoroid models of different cancer types for TME studies on day 30.    and immunofluorescence stainings (left, right) of a vascularized BrCa
Phase contrast (top), H&E (middle), and immunofluorescent stain-        tumoroid-on-a-chip model for studying the TME and drug sensibility
ing (bottom) of ALI PDTs for DAPI (purple) and VIM, CA9, S100           of cancer cells [186]. B. Copyright Elsevier, C. Copyright Wiley-VCH
or CK7, respectively (green) [183]. (C) Schematic depicting the gen-    GmbH, D. Copyright Royal Society of Chemistry. Reproduced with
eration of a bioprinted tumoroid co-culture model including fibro-      permission. Created with BioRender.com
blasts and endothelial cells (top) [184]. Immunofluorescent staining
artificial assembly of the isolated cells for direct co-culture         exposed to the atmosphere [183, 188] (Fig. 2A). ALI mod-
systems, Ootani et al. established a method to culture cells            els have already been established for several cancer types,
in their original environment by preserving the TME using               such as salivary gland cancer and kidney cancer (Fig. 2B).
ALI models [196]. Dissociated tissue is hereby directly                 This method leads to the differentiation and maturation of
embedded into an ECM such as collagen type I and/or a                   the cell types present in the tissue, resulting in organoids or
hydrogel, seeded onto a transwell mesh, and then cultured               tumoroids containing epithelial cells, as well as stromal and
in a way that only the basal part of the ECM or tissue is in            immune cells [13, 183, 197]. This way, PDTs can be cul-
contact with the culturing medium while the other part is               tured for months while mostly preserving the original TME,
13
A systematic review on the culture methods and applications of 3D tumoroids for cancer research and…
thus reflecting the physiological tumor site. However, the               how stroma cells could be targeted for anti-cancer therapies
limited availability of primary tumor tissue, the slow growth            [204].
of a tissue ALI culture, and the partial loss of stromal cells
during long-term culture, makes this method unsuitable for                  The importance of specific fibroblast subtypes and the
high-throughput analyses such as drug screens [198]. Tissue              synergistic cross-talk between the tumor and its TME for
ALI culture can nevertheless be a powerful direct co-culture             cancer progression is well understood. However, how tumor
model for studying processes like tumor progression, ther-               cells reprogram NFs into CAFs is still largely unknown. To
apy response to selected drugs, or the presence and infiltra-            investigate whether the mutational status of a tumor could
tion of immune cells in the original tumor tissue.                       reprogram and determine the fibroblast phenotype, Shaas-
                                                                         hua et al. analyzed the differences between NFs indirectly
2.2.1.1 Stromal cells As mentioned above, stromal cells in               co-cultured with either BRCA wild-type or germline BRCA
the TME are involved in many tumor promoting processes.                  mutated PaCa tumoroids [205]. Different fibroblast sub-
CAFs can accelerate tumor growth by synthesizing and                     types were indeed derived from the NFs depending on the
remodeling the ECM, and by producing growth factors that                 BRCA mutational status, thus suggesting that each tumor
induce angiogenesis. Additionally, by negatively affecting               might shape its individual TME.
drug access to the tumor they can influence therapy response                In summary, germline mutations can not only change
[187, 199–201]. We recently developed a PDT fibroblast                   the behavior of the tumor itself but can also influence the
co-culture model using matched tumoroids and CAFs or                     surrounding stromal cells [205]. Thus, to overcome stroma-
normal fibroblasts (NFs) isolated from CRC patients [188].               mediated resistance, therapies have to target multiple cell
Both NFs and CAFs were able to support the growth and                    types in the TME simultaneously, and personalized medi-
differentiation of tumoroids without the addition of specific            cine approaches are necessary for efficient treatment.
niche factors, suggesting a better reflection of the in vivo
tumor heterogeneity than conventional PDT cultures. In                   2.2.1.2 Leukocytes Immune cells are another important
addition, we observed an enhanced cancer-promoting phe-                  cell type in the TME inducing both anti- and pro-tumori-
notype of NFs upon co-culture with the tumoroids, high-                  genic effects, which represent promising targets for efficient
lighting the dynamic and interdependent cross-talk between               cancer treatment. Even though co-cultures of immortalized
the tumor and its microenvironment [188]. These co-culture               2D cell lines with immune cells, or animal models, already
models not only reflect the microenvironment but can also                led to the development of promising cancer treatments
be used to model the influence of the TME on cancer cell                 targeting the immune system, tumoroids co-cultured with
drug response. By testing the response of both human and                 immune cells might be another suitable model for testing
murine liver tumoroids to conventional anti-cancer drugs                 patient-specific responses to various immunotherapies [32,
such as regorafenib, sorafenib, and 5-FU, it was shown that              40, 206, 207].
the direct co-culture with CAFs or the addition of CAF-
conditioned medium to the liver tumoroids led to a higher                   Many groups have already established co-cultures of
resistance of the cancer cells [79]. Also, PaCa PDTs, which              tumoroids with lymphocytes focusing on T cells and natural
were directly co-cultured with CAFs, showed higher resis-                killer (NK) cells to improve the modeling of immunother-
tance against commonly used PaCa drugs when compared                     apies like CAR T cells and immune checkpoint blockade
to tumoroid-only cultures [187]. Single cell RNA-Sequenc-                [208]. CAR T cell therapy has proven successful for leu-
ing revealed differences in gene expression in the PDTs                  kemia, but models for investigating the cytotoxic effect
induced by the CAFs, which led to the induction of EMT                   of these cells on solid tumors like CRC are still needed.
in the cancer cells. The authors suggested that this EMT                 Therefore, Schnalzger et al. developed a CRC PDT co-cul-
induction contributed to the chemo-protectant effect seen in             ture model as a sensitive in vitro platform to study patient-
their co-culture system [187]. Similar EMT-inducing effects              specific treatment responses [207]. Other groups focused
have been seen in other studies where EMT was partially                  on investigating the mechanism of PD-1/PD-L1 immune
induced in cancer cells by direct co-culturing of CAFs and               checkpoint blockade using advanced co-culture models, and
human CRC tumoroids mimicking either early or late stage                 showed that human and murine lymphoma tumors and their
CRC [202, 203]. Further on, inhibition of WNT signaling in               TME could be cultivated for several weeks using an adapted
CAFs was shown to induce different CAF subtypes result-                  ALI model [183, 209]. The system reflected the physiologi-
ing in repression of EMT in CRC tumoroids, highlighting                  cal properties of the tissue, and cell composition including
                                                                         lymphoid cells and supporting T helper cells was preserved.
                                                                         Importantly, the ALI cultures functionally recapitulated
                                                                         PD-1/PD-L1 dependent immune checkpoint blockade,
                                                                                                                            13
                                                                                                                     J. Kalla et al.
highlighting the potential of this model to develop personal-    time consuming, and high-throughput methods still have
ized immunotherapies for lymphomas [183, 209].                   to be improved. Automated methods like bioprinting could
   Similarly, a recent human tumoroid and immune cell co-        help to overcome some of these limitations.
culture model for gastric cancer was developed [189]. In
this model, dendritic cells were first activated by a patient-   2.2.2 Bioprinting
derived tumor antigen, and then used to prime CD8 + T
cells. The authors suggest that the direct T cell activation     Bioprinting allows for the exact positioning of different
in their model is superior to activation by anti-CD28-coated     cell types and biomaterials in a 3D space with the help
plates, which was used in a similar co-culture model [210].      of a mechanical and computer-assisted system to mimic
   Moreover, focusing on immune checkpoint blockade              the in vivo spatial architecture of a tissue or tumor and its
therapies, novel bispecific antibodies for PD-1 and PD-L1        microenvironment [216]. The biggest advantage of using a
were used to restore NK and CD8 + T cell activity in ovar-       bioprinting-based approach to generate preclinical cancer
ian PDT co-cultures with intra-tumoral immune cells [211].       models is the standardization of cell dispensing [217], and
Immune cell reactivation was shown to be mediated by the         the possibility to construct an artificial 3D tumor including
partial downregulation of BRD1, and inhibition of BRD1           different cell types, structures, and ECMs for more precise
had a similar anti-tumoral effect in vitro and in vivo. Thus,    personalized medicine approaches [218].
BRD1 inhibition could provide a new therapy option for              Early studies focused on generating such models by
ovarian cancer patients to circumvent immune cell evasion        using bioprinted cancer cell lines or single cell suspensions
[211].                                                           [184]. Recently, the dispersion of organoids and tumoroids
   Myeloid immune cells such as tumor-associated macro-          together with stromal cells allowed for the development of
phages (TAMs) play an important role in the TME to sup-          co-culture models including the TME (Fig. 2C) [219].
port tumor growth, and thus represent important therapeutic         CRC microtissues were produced according to patient-
targets. Therefore, both conventional organoid [212] and         specific colonoscopy images by printing PDTs surrounded
ALI co-culture models combining tumoroids and macro-             by healthy organoids to model the interaction of a tumor
phages have been developed [213].                                with normal adjacent tissue [220]. As the in vitro treatment
   By including multiple immune cell types in one co-            response of these microtissues to the standard 5-FU therapy
culture model, it is possible to further analyze the complex     reflected the patient response, the model could be used as a
interplay between the tumor, and between different kinds of      more physiological drug screening platform. Additionally,
immune cells. This way, it was shown that reprogramming          the patient-specific risk of tumor invasion into the surround-
of TAMs following monoamine oxidase A inhibitor (MAO-            ing tissues was calculated in this model by correlating the
Ai) treatment, a widely used class of drugs for the treatment    number and distance of invading tumor cells. This could
of depression and Parkinson’s disease, enabled the induc-        provide a real-time quantitative readout for analyzing can-
tion of anti-tumor T cell reactivity in a melanoma co-culture    cer progression and metastasis [220].
model [214]. MAO-Ai could thus be used in combination               In another recent study, microtissues consisting of
with PD-1/PD-L1 inhibitors to reactivate the anti-tumor          patient-derived lung tumoroids in co-culture with matched
immune response as a new potential treatment for several         CAFs and endothelial cells were generated [221]. After
cancer types.                                                    printing the vessel structures and seeding the CAFs, the
   However, co-cultures can not only be used to study the        tumoroids suspended in a hydrogel derived from por-
interactions between tumoroids and multiple immune cells         cine lung tissue were printed into the same compartment.
but also to analyze more complex interactions between cells      An active fusion of the stromal cells and tumoroids was
in the TME, including tumor cells, immune cells, and stro-       observed, and microvessels formed to directly interact with
mal cells. For this, Sufi et al. developed the Thiol-reactive    the other cell types. After administering the drug poziotinib
Organoid Barcoding in situ Mass Cytometry (TOBis MC)             through the vessel structures it was seen that both the endo-
protocol, which allows for the analysis of organoid and          thelial cells and CAFs, but also the CAF-secreted matrix,
tumoroid lines in co-culture with leukocytes and fibroblasts     protected the lung tumoroids from the treatment. Thus, this
on single cell level to study the interactions between TAMs      model could be used to further test the influence of cell-cell
and CAFs in high-resolution [215].                               or cell-matrix interactions on the efficacy of drug delivery to
   In summary, organoid and tumoroid models in co-cul-           the tumor tissue [221].
ture with other cell types can be used to model the complex         Apart from printing models mimicking the TME, bio-
interactions of the TME. Even though the benefit of these        printing can be used for standardized cell dispensing for
co-culture models for improving or finding novel treatment       high-throughput applications. However, one drawback
options has been proven, the assembly of these models is         is the difficulty of printing bioink into small wells as the
13
A systematic review on the culture methods and applications of 3D tumoroids for cancer research and…
utilized ECMs can spread in the well and thus do not main-               treatment. Aside from its suitability for physiological drug
tain the shape necessary for 3D cell growth. To circumvent               testing, this chip design could be a useful tool to study and
the spreading, patient-derived glioblastoma or sarcoma cells             easily visualize differences in angiogenesis, proliferation,
were mixed with a bioink consisting of hyaluronic acid as                and migration [186]. Importantly, it has already been shown
well as collagen, and then printed into wells coated with                before that fluidic devices provide a more physiological
gelatin, which was later removed and substituted with                    model to study cancer cell migration [229].
medium [222]. In addition, acoustic bioprinting of small                    Using microfluidic devices with tumoroids alone, recent
droplets onto a hydrophobic substrate was used to generate               papers already described the advantages of chip-based mod-
BlCa-derived tumoroids consisting of both cancer cells and               els for a personalized medicine approach and more rep-
CAFs [223]. This method allows for the generation of large               resentative drug testing compared to static models. Both
numbers of uniform tumoroids that mimic the TME, which                   Mazzocchi et al. [143] and Jung et al. [230] used PDTs to
can easily be dispensed into small wells and be used in high-            perform medium-throughput drug screens for mesothelioma
throughput drug screens for personalized therapy.                        or small cell lung cancer (SCLC) patients, respectively. Drug
   Thus, the combination of 3D cell models and bioprinting               response to specific chemotherapeutic therapies was differ-
could help to facilitate reproducible drug testing, and the              ent between the tumoroid lines with different mutational
investigation of cellular processes in a 3D space with mul-              backgrounds but correlated well with the patient response.
tiple cell types and structures. However, these printed mod-             Importantly, SCLC tumoroids on the chip maintained a che-
els are still static and do not reflect the effect of mechanical         motherapy resistant core, which might be due to insufficient
forces like dynamic flow, or chemical gradients, which                   drug delivery also observed in in vivo tumors [230].
influence the tumor cell phenotype in vivo [224].                           To facilitate the translation of this drug screening method
                                                                         to the clinics, Schuster et al. developed an automated high-
2.2.3 Fluidic devices                                                    throughput screening platform on a chip [231]. The authors
                                                                         constructed a microfluidic system with which 20 indepen-
The important physical and chemical characteristics of the               dent experimental conditions could be tested on up to 10
TME can be modeled with microfluidic systems, where                      different patient-derived 3D cell lines. A customizable soft-
cells are cultured in chambers connected to microchannels                ware allows for the stimulation, assaying, and imaging of
supplying oxygen, growth factors, or drugs via dynamic                   the tumoroids without human intervention. Interestingly,
perfusion [225–227] (Fig. 2D). In a proof of concept study,              combinatorial treatments and time-dependent adminis-
CRC PDTs cultured on fluidic devices showed a higher via-                tration with multiple rounds of treatment showed greater
bility, proliferation rate, and tumoroid formation efficiency,           therapy efficacy on PaCa tumoroids compared to single
compared to tumoroids cultured in static ECM drops on a                  treatments [231].
plate [228]. Additionally, chip-grown tumoroids were big-                   To investigate the influence of the tumor stroma and the
ger in size and showed a multilayered morphology with                    method of drug delivery on treatment efficiency, a fluidic
crypt-like projections similar to the human colon, while                 system mimicking the TME for drug testing on PaCa PDTs
the conventional tumoroids formed more cystic monocel-                   has recently been established [232]. The vasculature was
lular structures. Interestingly, when testing the response of            modeled by growing endothelial cells in a perfusable micro-
PDTs to 5-FU no significant differences were seen between                fluidic scaffold. Tumoroids co-cultured with fibroblasts,
the culturing methods, suggesting that chip-grown 3D cells               which resulted in increased proliferation of cancer cells and
maintain their phenotype, while having a growth advantage,               enhanced tissue stiffness, were then added into adjacent
and thus validating the use of fluidic devices for tumoroid              compartments. When administering the chemotherapeutic
culture and disease modeling [228].                                      drug gemcitabine to the co-culture via diffusion through
   However, the tumor niche consists of multiple cell types,             the vessel-like structures, the drug had a reduced efficiency
and an extensive vasculature, through which nutrients, oxy-              compared to the administration to tumoroid single cultures
gen or drugs get delivered to the cancer cells. In order to              or to static co-cultures. Thus, the authors were able to reca-
mimic this vascular transport of factors and metabolites to              pitulate the microenvironment of a vascularized pancreatic
the tumor, endothelial cells together with fibroblasts were              tumor and to demonstrate the inhibitory effect of the vascu-
grown in microfluidic chambers to produce a 3D micro-                    lature cell barrier and the stroma on drug efficiency [232].
vascular network, before adding primary BrCa tumoroid-                      Interestingly, another PaCa-on-a-Chip model was used
like structures into a separate adjacent compartment [186].              to show that stroma-targeting agents do not influence the
Using this setup, vessel outgrowth towards the tumor cell                cell viability of tumoroids in monoculture, but lead to a sig-
chamber was observed in addition to tumor cells invading                 nificant increase of chemotherapeutic anti-cancer effects in
the vasculature chamber after inducing EMT via TGF-β                     a fluidic device co-culture [233]. Using a similar model, it
                                                                                                                            13
                                                                                                                                  J. Kalla et al.
was reported that different drugs show different efficacies in           3 In vivo methods and applications
targeting PaCa cells in normoxic versus hypoxic conditions
[234]. Thus, both publications highlight the importance of               3.1 Modeling of tumor biology
including cells of the TME and relevant oxygen levels for
microfluidic drug response studies.                                      Even though tumoroid models can deliver meaningful trans-
   In conclusion, tumoroid-on-a-chip models provide a                    latable results for basic cancer research and patient therapy
powerful tool to investigate biological processes and to                 options, the assessment of systemic effects and the influence
assess drug responses in an efficient and more physiological             of a more complex in vivo TME can only be validated in ani-
manner compared to static tumoroid cultures.                             mal models [51] (Fig. 3A). In the past, whole tissue pieces,
                                                                         digested primary tissue, or 2D cell lines were grafted into
Fig. 3 In vivo methods for tumoroid models. (A) Overview of in vivo      treatment of patient-derived xenograft mice representing a pretreat-
applications of both human and murine tumoroids. (B) Light micros-       ment tumor with either patient-matched neoadjuvant therapy (includes
copy pictures of tumoroids with different phenotypes (Scale bar          the drugs doxorubicin hydrochloride (Adriamycin) and cyclophospha-
200 μm) and H&E staining of corresponding tumors after re-inoculation    mide, followed by treatment with paclitaxel (Taxol) = AC-T) (left) or
of subcutaneous tumor-derived tumoroids (Scale bar 50 μm) [237]. (C)     with drugs selected from a xenograft-derived tumoroid screen (right)
Pearson correlation coefficient plots of gene expression between pros-   [108]. B, C, E adapted from corresponding citations; Springer Nature
tate tumors and matched tumoroid lines [238]. (D) Dynamic changes        (Creative Commons Attribution 4.0 International). D adapted from
of immune cell populations during early, middle, and late stages in      Wiley (Creative Commons Attribution 4.0 International). Created with
a novel tumoroid-based liver metastasis model [239]. (E) Real-time       BioRender.com
13
A systematic review on the culture methods and applications of 3D tumoroids for cancer research and…
animals as allografts (between same species) or xenografts               heterogeneity and plasticity hindering efficient treatment,
(between different species) either subcutaneously or ortho-              metastasis formation is one of the leading causes of cancer-
topically [235]. These models have lately been expanded                  related deaths [242]. CTCs can spread to distant secondary
to transplant organoid and tumoroid lines into animals as                sites to induce metastasis [243]. As CTCs are a rare and het-
allografts and xenografts. Importantly, the pathological and             erogeneous cell population, De Angelis et al. established a
invasive features of the original tumor and drug response of             model to study the biological features and possible drug sen-
the patient are retained in these models, providing a suitable           sitivities of these cells [244]. The group derived tumoroids
alternative to investigate mechanisms of malignant transfor-             from both orthotopic CRC xenografts and CTCs isolated
mation, tumor plasticity, and metastasis in vivo [236].                  from the blood of the same mice. As expected, CTC-derived
                                                                         tumoroids showed a more aggressive and migratory pheno-
3.1.1 Cancer initiation and progression                                  type in addition to higher stem cell and EMT marker expres-
                                                                         sion compared to the xenograft-derived tumoroids. The
To study the tumorigenic potential of cancer cells based                 authors also showed that CTC-derived tumoroids are highly
on common mutational patterns specific for endometrial                   sensitive to drugs targeting the Survivin pathway including
cancer, murine tumoroids that overexpress KrasG12D and                   YM155 and quercetin, providing a new possible treatment
carry genetic deletions of various tumor suppressor genes                to inhibit metastasis formation. As this model reflected the
(Pten, Tp53) were allografted subcutaneously into nude                   properties of CTCs isolated from CRC patients, De Angelis
mice [237]. Different mutational backgrounds led to the                  et al. hypothesized that in the future patient-specific drug
development of specific tumor subtypes with correspond-                  sensitivities could be studied in a metastasis xenograft
ing tumoroids having an either more cystic or spindle-like               model established from the patient’s primary tumor even
morphology because of an irreversible EMT during trans-                  before the onset of metastatic disease, and thus prevent
formation. This also correlated with a less or more aggres-              tumor progression [244].
sive phenotype, respectively [237] (Fig. 3B).                                The same group also developed another preclinical
   As the specific cancer subtype also plays an important                metastasis model for CRC [245]. After orthotopically graft-
role in the aggressiveness and treatment of PaCa, Miya-                  ing patient-derived cells into immunodeficient mice, the
bayashi et al. established an in vivo model to investigate the           authors generated tumoroids from the primary tumor site
role of patient-specific genetic and epigenetic aberrations              and from spontaneous liver metastases and analyzed the
for PaCa heterogeneity [240]. The authors derived tumor-                 EMT state together with the drug response. As expected,
oids from patients and xenografted them orthotopically into              metastatic tumoroids displayed a more mesenchymal phe-
the pancreatic ducts of immunodeficient mice. Tumors were                notype and increased chemoresistance. Interestingly, high
either indolent with a luminal morphology or progressed                  numbers of CTCs in patients correlated with an increased
to a more aggressive basal-like subtype based on epigen-                 engraftment efficiency of the corresponding tumor-derived
etic aberrations and deregulation of KRAS. In contrast to                cells, again highlighting the important role of CTCs for can-
Maru et al. [237], this group argued that cellular plasticity            cer progression [245].
is mainly influenced by the stroma and microenvironment                      EMT is not only an important characteristic of CTCs, but
rather than cellular clonality, thus suggesting that allo- and           also of the cancer cells in tumor lesions. Recently it was
xenografts should be performed orthotopically to recapitu-               shown that hybrid cells, presenting both epithelial and mes-
late the corresponding tumor niche for translatable results              enchymal characteristics, can be found in human tumor tis-
[240].                                                                   sues, GEMMs, and tumoroids of BrCa [246]. The number of
                                                                         these cells correlated with worse overall survival in patients,
3.1.2 Cellular plasticity and metastasis                                 while driving invasion in vitro. Using tail vein injection or
                                                                         orthotopic engraftment of tumor cells or tumoroids, respec-
Cellular plasticity and clonality can not only be influenced             tively, heterogeneous EMT states were identified within
by the microenvironment, but also by specific therapies                  the same tumor. Sequential molecular EMT programs were
[241]. To investigate resistance mechanisms of androgen                  essential for tumor cell invasion and colonization during the
pathway directed therapy, Lee et al. generated tumoroids                 metastatic process. Thus, both models could be used to fur-
from patient-derived xenograft models of bone metastatic                 ther investigate the influence of the EMT status on patient
PCa. Androgen ablation led to the development of revers-                 survival [246].
ibly dormant basal-luminal-like hybrid cells, highlighting                   To further understand the mechanism of metastasis for-
how some cancer therapies might even support the pro-                    mation, the role of the immune system in the metastatic
gression of the disease, thus suggesting that treatment regi-            niche was also recently investigated in a PCa model of liver
mens might have to be reviewed [241]. Apart from tumor                   metastasis [239]. Murine tumoroids generated from GEMMs
                                                                                                                             13
                                                                                                                     J. Kalla et al.
with PCa specific mutations were orthotopically allografted      case study showed that drug testing using xenografted PDTs
into immunocompetent mice, which resulted in metastatic          is feasible in real time and can lead to beneficial results
spread to the liver after only 30 days. This model is not        for patients [108]. Drug screens with extensive compound
only less time consuming compared to original GEMMs              libraries can also help to further elucidate the mechanism
and xenografts, and more reliable than patient-derived cell      of cancer progression. Even though many epigenetic aber-
lines, but importantly also allows for the investigation of      rations play a role in the progression of BlCa, the use of
the immune cells in the metastatic niche. The authors found      epigenetic drugs as a potential treatment has so far only
that an immune suppressive microenvironment mediated             been investigated in five clinical trials [249]. The histone
mainly by neutrophils, anti-inflammatory macrophages, and        deacetylase activator SRT1720, which specifically targets
CD4 + T cells was essential for the survival of tumor cells      SIRT1, was recently identified as a potent drug to inhibit
and progression of metastases (Fig. 3D). This model might        the growth of both BlCa tumoroids and xenografts [249].
be relevant for testing drugs targeting immune cells, and for    Mechanistically, SIRT1 activation resulted in deacetylation
elucidating cell-cell crosstalk in the metastatic niche [239].   of HIF1α and subsequent repression of the hypoxia path-
   In summary, organoids and tumoroids can be used in            way. This pathway has been correlated with poor prognosis
an in vivo setting to investigate many aspects of cancer         in patients, highlighting the potential of epigenetic therapies
development and progression, highlighting the advantages         for BlCa [249]. In contrast, SIRT1 was also reported to pro-
of orthotopic grafts and allografts. Thus, these models are      mote the progression of BlCa [250, 251]. However, these
highly suitable for drug testing and the development of          studies were performed on 2D cell lines and might thus be
novel therapies.                                                 less representative compared to 3D models and xenografts
                                                                 [249].
3.2 Therapy development and drug testing in vivo                    Apart from mouse models, rats are a widely used animal
                                                                 model for cancer research and treatment development. By
Patient-derived in vitro models in combination with matched      orthotopically transplanting kidney tumoroids derived from
in vivo models represent a suitable model system for per-        patient-specific iPSCs into rats, a fully vascularized xeno-
sonalized medicine applications. Multiple groups have used       graft model for angiomyolipoma, a rare kidney tumor, was
this approach to study cancer subtypes, patient-specific         generated [252]. Using this model, the therapeutic efficacy
drug responses, and to identify potential alternative thera-     of mTOR inhibition by rapamycin-loaded nanoparticles, the
pies. Importantly, while both 3D in vitro and in vivo models     main treatment for angiomyolipoma, was confirmed, sug-
retain the genetic and epigenetic background of the primary      gesting that this model would also be suitable to identify
tumor (Fig. 3C), xenograft-derived 2D cell lines can acquire     novel alternative treatments for this rare cancer type [252].
additional mutations in culture, suggesting that 3D culture         Additionally, zebrafish models are well established to
systems are more stable and representative models for drug       study tumor metastasis and are also commonly used for in
screening [247].                                                 vivo drug testing [253]. A comparative analysis of relapsed
   Several recent studies provided evidence for the usabil-      pediatric malignancies used patient-derived spheroid cul-
ity of PDTs for different tumor types, including endometrial     tures, tumor cells isolated from corresponding mouse
cancer [248], glioma [247], BrCa [108], and metastatic PCa       xenografts, long-term organoid-like cultures, and zebrafish
[238]. These “living” biobanks reflected patient-specific        xenografts to compare drug responses in the different model
drug response, mimicked tumor metastasis [248], and were         systems. The zebrafish model proved to be a promising addi-
used to identify alternative treatment options such as the       tion to current drug testing tools to rapidly assess potential
chemotherapeutic dianhydrogalactitol (VAL-083) for gli-          treatments in vivo, and could be included for personalized
oma [247], or multi-kinase inhibitors for therapy resistant      medicine approaches in the future [253].
metastatic PCa [238].                                               In summary, a variety of different animal- and patient-
   Additionally, Guillen et al. reported a first proof that      derived in vitro and in vivo models are available, which can
results generated by patient-specific tumoroids in vitro could   be tailored to answer specific questions for basic and trans-
be translated to the clinic [108] (Fig. 3E). The group had       lational research.
already generated a xenograft model with a matched tumor-
oid line of a BrCa patient and performed a drug screen iden-
tifying eribulin as a novel drug that showed effective growth
inhibition in vitro and complete regression with no recur-
rence in the mice. The patient went into complete remission
after being treated with the same drug, however due to other
complications later unfortunately died. Nonetheless, this
13
A systematic review on the culture methods and applications of 3D tumoroids for cancer research and…
                                                                                                                             13
                                                                                                                            J. Kalla et al.
for assessing the complex biological effects of drugs, which       have already been established by the National Cancer Insti-
cannot yet be fully replaced by PDT models.                        tute in the US [289], and by a consortium of several Euro-
                                                                   pean institutions (Human cancer models initiative) [290].
                                                                   Recently, the Organoid Cell Atlas, which in addition to
5 Conclusion and outlook                                           tumor samples and tumoroids also includes healthy organ-
                                                                   oids, was launched as a comprehensive database [291, 292].
As cancer is a highly heterogeneous disease, versatile             Similarly, OrganoidDB has already collected bulk and sin-
models that can easily be manipulated in vitro, and stably         gle cell transcriptomic data of more than 16.000 organoid
reflect the characteristics of the primary tumor, are needed.      and tumoroid models for both human and mouse samples, in
Thus, tumoroids have a great potential to replace standard         addition to data generated from primary tissue and cell lines
2D cell culture or animal models. However, to adequately           for comparison [293].
model the inter- and intra-patient heterogeneity, several             Lastly, 3D cell models are a promising tool to reduce the
tumoroid lines would have to be generated from the same            number of animals used for preclinical cancer research in
tissue biopsy, which is not the standard practice [162, 163].      concordance with the 3R principles [294]. Due to a recent
Additionally, limitations regarding tumoroid cultivation           ground-breaking new law, the US Food and Drug Adminis-
methods still need to be overcome. For example, artificial         tration can now approve drugs for clinical trials on humans
ECM hydrogels [257, 273, 274], or human-derived ECMs               that were tested only in vitro on organ-on-a-chip models or
[275], have been developed to limit negative cross-species         organoid and tumoroid models [295]. Although alternative
interactions for PDTs. Additionally, cultures with no matrix       methods for drug testing, such as 3D cell models, are not
at all [276], or bioreactors [277] could be used for tumor-        yet standard practice, they hold great promise for redefin-
oid expansion and drug screens in the future. Importantly,         ing preclinical cancer research and precision medicine in the
many groups have focused on the automation of 3D cell              future, due to their molecular and phenotypic similarities to
culturing and analysis methods for high-throughput appli-          patient tumors.
cations to achieve a better translation of results to the clinic
[278–280]. For high-throughput drug screens, protocols             Acknowledgements The authors want to thank Milena Mijović, Kata-
                                                                   rina Mišura, and Luisa Grader for compiling references. The authors
that either use tumoroid-derived single cells, or small- to        also want to thank Maša Zrimšek, Kristina Draganić, Katarina Mišura,
medium-sized tumoroids in suspension, have already been            and Gabriel Wasinger for providing high-level feedback on the draft.
developed [219]. Additionally, bioinformatic approaches
and mathematical modeling have been combined with 3D               Author contributions Writing - original draft: JK, JP, TM, LT; Writ-
cancer models to test for drug sensitivities faster [281–283].     ing - reviewing and editing: JK, JP, GE; Conceptualization: JK, JP,
                                                                   GE; Visualization: JP, JK; Supervision: GE. All authors have read and
To further improve the model systems available for devel-          approved the final manuscript.
oping and testing novel treatment options, multi-organ chip-
based systems were developed, which are used to study the          Funding This work was supported by the Austrian Science Founda-
systemic effects of anti-cancer drugs on healthy tissue or         tion (FWF) project P 32771 (Gerda Egger, Jessica Kalla); Austrian
other organs [284, 285], and thus could reduce the use of          Science Foundation (FWF) SFB F83 (Gerda Egger, Janette Pfneissl);
                                                                   Austrian Science Foundation (FWF), doc.funds grant DOC59 (Gerda
animal models for drug testing.                                    Egger, Theresia Mair); City of Vienna Fund for Innovative Interdisci-
   In the future, living biobanks of organoid and tumoroid         plinary Cancer Research, 21118 (Gerda Egger); City of Vienna Fund
models will be essential for real-time personalized treat-         for Innovative Interdisciplinary Cancer Research, 21209 (Loan Tran);
ment of cancer patients, as summarized in a recent review          Austrian Academy of Sciences, Doc Fellowship 25276 (Loan Tran).
                                                                   Open access funding provided by Medical University of Vienna.
[286]. A number of clinical trials are already investigating
the outcome of personalized therapy selection based on             Data availability No datasets were generated or analysed during the
tumoroid drug sensitivity testing for several cancer types,        current study.
including PaCa (NCT04931394, NCT04931381), BrCa
(NCT04450706, NCT03544047, NCT05177432), lung                      Declarations
cancer (NCT05136014), BlCa (NCT05024734), and sev-
eral different cancers (NCT04279509) [287]. However, the           Ethics approval and consent to participate Not applicable.
standardization of methods has to be addressed by regula-
                                                                   Copyright Where specified, figures were reproduced from cited pub-
tory authorities and put into practice in the industry to suc-     lications with permission of the respective journals, or were adapted
cessfully implement 3D models in the clinics. Open data            from cited publications under the Creative Commons Attribution 4.0
platforms that include repositories of protocols, donors,          International (https://creativecommons.org/licenses/by/4.0/.
organoids and tumoroids, and ethical information, are
needed [288]. Such biobanks for human-derived 3D models            Consent for publication Not applicable.
13
A systematic review on the culture methods and applications of 3D tumoroids for cancer research and…
Competing interests The authors declare that they have no competing        12. M. Huch, B.-K. Koo, Modeling mouse and human development
interests.                                                                     using organoid cultures. Development. 142, 3113–3125 (2015).
                                                                               https://doi.org/10.1242/dev.118570
Open Access This article is licensed under a Creative Commons              13. X. Li, L. Nadauld, A. Ootani, D.C. Corney, R.K. Pai, O. Gevaert
Attribution 4.0 International License, which permits use, sharing,             et al., Oncogenic transformation of diverse gastrointestinal tis-
adaptation, distribution and reproduction in any medium or format,             sues in primary organoid culture. Nat. Med. 20, 769–777 (2014).
as long as you give appropriate credit to the original author(s) and the       https://doi.org/10.1038/nm.3585
source, provide a link to the Creative Commons licence, and indicate       14. T. Sato, R.G. Vries, H.J. Snippert, van de M. Wetering, N. Barker,
if changes were made. The images or other third party material in this         D.E. Stange et al., Single Lgr5 stem cells build crypt-villus struc-
article are included in the article’s Creative Commons licence, unless         tures in vitro without a mesenchymal niche. Nature. 459, 262–
indicated otherwise in a credit line to the material. If material is not       265 (2009). https://doi.org/10.1038/nature07935
included in the article’s Creative Commons licence and your intended       15. C. Corrò, L. Novellasdemunt, V.S.W. Li, A brief history of organ-
use is not permitted by statutory regulation or exceeds the permitted          oids. Am. J. Physiology-Cell Physiol. 319, C151–C165 (2020).
use, you will need to obtain permission directly from the copyright            https://doi.org/10.1152/ajpcell.00120.2020
holder. To view a copy of this licence, visit http://creativecommons.      16. S. Yang, H. Hu, H. Kung, R. Zou, Y. Dai, Y. Hu et al., Organ-
org/licenses/by/4.0/.                                                          oids: The current status and biomedical applications. MedComm
                                                                               (2020) 2023;4:e274. https://doi.org/10.1002/mco2.274
                                                                           17. E. Driehuis, K. Kretzschmar, H. Clevers, Establishment of
                                                                               patient-derived cancer organoids for drug-screening applications.
References                                                                     Nat. Protoc. 15, 3380–3409 (2020). https://doi.org/10.1038/
                                                                               s41596-020-0379-4
1.  K. Ronaldson-Bouchard, I. Baldassarri, D.N. Tavakol, P.L.              18. J. Drost, W.R. Karthaus, D. Gao, E. Driehuis, C.L. Sawyers, Y.
    Graney, M. Samaritano, E. Cimetta et al., Engineering complex-             Chen et al., Organoid culture systems for prostate epithelial tis-
    ity in human tissue models of cancer. Adv. Drug Deliv. Rev. 184,           sue and prostate cancer tissue. Nat. Protoc. 11, 347–358 (2016).
    114181 (2022). https://doi.org/10.1016/j.addr.2022.114181                  https://doi.org/10.1038/nprot.2016.006
2. H. Sajjad, S. Imtiaz, T. Noor, Y.H. Siddiqui, A. Sajjad, M. Zia,        19. S. Nuciforo, I. Fofana, M.S. Matter, T. Blumer, D. Calabrese,
    Cancer models in preclinical research: a chronicle review of               T. Boldanova et al., Organoid models of Human Liver cancers
    advancement in effective cancer research. Anim. Model. Exp.                derived from Tumor needle biopsies. Cell. Rep. 24, 1363–1376
    Med. 4, 87–103 (2021). https://doi.org/10.1002/ame2.12165                  (2018). https://doi.org/10.1016/j.celrep.2018.07.001
3. K. Duval, H. Grover, L.-H. Han, Y. Mou, A.F. Pegoraro, J. Fred-         20. C.J. Phifer, K.N. Bergdorf, M.E. Bechard, A. Vilgelm, N. Bare-
    berg et al., Modeling physiological events in 2D vs. 3D cell               gamian, O.G. McDonald et al., Obtaining patient-derived cancer
    culture. Physiol. (Bethesda). 32, 266–277 (2017). https://doi.             organoid cultures via fine-needle aspiration. STAR. Protocols. 2,
    org/10.1152/physiol.00036.2016                                             100220 (2021). https://doi.org/10.1016/j.xpro.2020.100220
4. M. Kapałczyńska, T. Kolenda, W. Przybyła, M. Zajączkowska,              21. A.E. Vilgelm, K. Bergdorf, M. Wolf, V. Bharti, R. Shattuck-
    A. Teresiak, V. Filas et al., 2D and 3D cell cultures– a compari-          Brandt, A. Blevins et al., Fine-needle aspiration-based patient-
    son of different types of cancer cell cultures. Arch. Med. Sci. 14,        derived cancer organoids. iScience. 23, 101408 (2020). https://
    910–919 (2018). https://doi.org/10.5114/aoms.2016.63743                    doi.org/10.1016/j.isci.2020.101408
5. J.-P. Gillet, A.M. Calcagno, S. Varma, M. Marino, L.J. Green,           22. F. Schutgens, M.B. Rookmaaker, T. Margaritis, A. Rios, C.
    M.I. Vora et al., Redefining the relevance of established cancer           Ammerlaan, J. Jansen et al., Tubuloids derived from human
    cell lines to the study of mechanisms of clinical anti-cancer drug         adult kidney and urine for personalized disease modeling.
    resistance. Proc Natl Acad Sci. 108, 18708–13 (2011). https://doi.         Nat. Biotechnol. 37, 303–313 (2019). https://doi.org/10.1038/
    org/10.1073/pnas.1111840108                                                s41587-019-0048-8
6. L. Trastulla, J. Noorbakhsh, F. Vazquez, J. McFarland, F. Iorio,        23. O. Kopper, de C.J. Witte, K. Lõhmussaar, J.E. Valle-Inclan, N.
    Computational estimation of quality and clinical relevance of              Hami, L. Kester et al., An organoid platform for ovarian cancer
    cancer cell lines. Mol. Syst. Biol. 18, e11017 (2022). https://doi.        captures intra- and interpatient heterogeneity. Nat. Med. 25, 838–
    org/10.15252/msb.202211017                                                 849 (2019). https://doi.org/10.1038/s41591-019-0422-6
7. S. Abdolahi, Z. Ghazvinian, S. Muhammadnejad, M. Saleh,                 24. N. Sachs, A. Papaspyropoulos, D.D. Zomer-van Ommen, I. Heo,
    H. Asadzadeh Aghdaei, K. Baghaei, Patient-derived xeno-                    L. Böttinger, D. Klay et al., Long-term expanding human airway
    graft (PDX) models, applications and challenges in cancer                  organoids for disease modeling. EMBO J. 38, e100300 (2019).
    research. J. Transl Med. 20, 206 (2022). https://doi.org/10.1186/          https://doi.org/10.15252/embj.2018100300
    s12967-022-03405-8                                                     25. K. Kinoshita, Y. Tsukamoto, Y. Hirashita, T. Fuchino, S. Kurogi,
8. K.F. Idrisova, H.-U. Simon, M.O. Gomzikova, Role of patient-                T. Uchida et al., Efficient establishment of bile-derived organoids
    derived models of cancer in translational oncology. Cancers. 15,           from biliary cancer patients. Lab. Invest. 103, 100105 (2023).
    139 (2023). https://doi.org/10.3390/cancers15010139                        https://doi.org/10.1016/j.labinv.2023.100105
9. K.H. Griffin, S.W. Fok, J. Kent Leach, Strategies to capitalize         26. D. Gao, I. Vela, A. Sboner, P.J. Iaquinta, W.R. Karthaus, A.
    on cell spheroid therapeutic potential for tissue repair and dis-          Gopalan et al., Organoid cultures derived from patients with
    ease modeling. Npj Regen Med. 7, 1–13 (2022). https://doi.                 advanced prostate cancer. Cell. 159, 176–187 (2014). https://doi.
    org/10.1038/s41536-022-00266-z                                             org/10.1016/j.cell.2014.08.016
10. M.A. Lancaster, M. Huch, Disease modelling in human organ-             27. H.K. Kleinman, G.R. Martin, matrigel, basement membrane
    oids. Dis. Model. Mech. 12, dmm039347 (2019). https://doi.                 matrix with biological activity. Sem. Cancer Biol. 15, 378–386
    org/10.1242/dmm.039347                                                     (2005). https://doi.org/10.1016/j.semcancer.2005.05.004
11. T. Sato, D.E. Stange, M. Ferrante, R.G.J. Vries, Van J.H. Es, Van      28. Z. Gan, X. Qin, H. Liu, J. Liu, J. Qin, Recent advances in defined
    den S. Brink et al., Long-term expansion of epithelial organoids           hydrogels in organoid research. Bioactive Mater. 28, 386 (2023).
    from human colon, adenoma, adenocarcinoma, and Barrett’s epi-              https://doi.org/10.1016/j.bioactmat.2023.06.004
    thelium. Gastroenterology. 141, 1762–1772 (2011). https://doi.         29. M. Fujii, M. Matano, K. Nanki, T. Sato, Efficient genetic engi-
    org/10.1053/j.gastro.2011.07.050                                           neering of human intestinal organoids using electroporation.
                                                                                                                                      13
                                                                                                                                       J. Kalla et al.
      Nat. Protoc. 10, 1474–1485 (2015). https://doi.org/10.1038/                EMBO Mol. Med. 14, e16001 (2022). https://doi.org/10.15252/
      nprot.2015.088                                                             emmm.202216001
30.   J.F. Dekkers, van E.J. Vliet, N. Sachs, J.M. Rosenbluth, O.          46.   T. Wakamatsu, H. Ogawa, K. Yoshida, Y. Matsuoka, K. Shizuma,
      Kopper, H.G. Rebel et al., Long-term culture, genetic manipu-              Y. Imura et al., Establishment of Organoids from Human Epi-
      lation and xenotransplantation of human normal and breast can-             thelioid Sarcoma with the air-liquid interface organoid cultures.
      cer organoids. Nat. Protoc. 16, 1936–1965 (2021). https://doi.             Frontiers in Oncology 2022;12.
      org/10.1038/s41596-020-00474-1                                       47.   C. Calandrini, van S.R. Hooff, I. Paassen, D. Ayyildiz, S. Dera-
31.   J. Clinton, P. McWilliams-Koeppen, Initiation, expansion, and              khshan, M.E.M. Dolman et al., Organoid-based drug screening
      cryopreservation of human primary tissue-derived normal and                reveals neddylation as therapeutic target for malignant rhabdoid
      diseased organoids in embedded three-dimensional culture. Curr.            tumors. Cell. Rep. 36, 109568 (2021). https://doi.org/10.1016/j.
      Protocols Cell. Biol. 82, e66 (2019). https://doi.org/10.1002/             celrep.2021.109568
      cpcb.66                                                              48.   D. Dutta, I. Heo, H. Clevers, Disease modeling in stem cell-
32.   K. Yuki, N. Cheng, M. Nakano, C.J. Kuo, Organoid models                    derived 3D organoid systems. Trends Mol. Med. 23, 393–410
      of tumor immunology. Trends Immunol. 41, 652–664 (2020).                   (2017). https://doi.org/10.1016/j.molmed.2017.02.007
      https://doi.org/10.1016/j.it.2020.06.010                             49.   I. Vitale, E. Shema, S. Loi, L. Galluzzi, Intratumoral hetero-
33.   J. Drost, H. Clevers, Organoids in cancer research. Nat.                   geneity in cancer progression and response to immunother-
      Rev. Cancer. 18, 407–418 (2018). https://doi.org/10.1038/                  apy. Nat. Med. 27, 212–224 (2021). https://doi.org/10.1038/
      s41568-018-0007-6                                                          s41591-021-01233-9
34.   T. Seidlitz, S.R. Merker, A. Rothe, F. Zakrzewski, C. von Neu-       50.   I. Bozic, T. Antal, H. Ohtsuki, H. Carter, D. Kim, S. Chen et al.,
      beck, K. Grützmann et al., Human gastric cancer modelling using            Accumulation of driver and passenger mutations during tumor
      organoids. Gut. 68, 207–217 (2019). https://doi.org/10.1136/               progression. Proc Natl Acad Sci. 107, 18545–50 (2010). https://
      gutjnl-2017-314549                                                         doi.org/10.1073/pnas.1010978107
35.   R. Joshi, M. Castro De Moura, D. Piñeyro, D. Alvarez-Errico, C.      51.   J. Kim, B.-K. Koo, J.A. Knoblich, Human organoids: model sys-
      Arribas, M. Esteller, The DNA methylation landscape of human               tems for human biology and medicine. Nat. Rev. Mol. Cell. Biol.
      cancer organoids available at the American type culture collec-            21, 571–584 (2020). https://doi.org/10.1038/s41580-020-0259-3
      tion. Epigenetics n d;15:1167–1177. https://doi.org/10.1080/155      52.   J. Du, L. Fang, J. Zhao, Y. Yu, Z. Feng, Y. Wang et al., Zymosan-A
      92294.2020.1762398                                                         promotes the regeneration of intestinal stem cells by upregulating
36.   H. Clevers, Modeling Development and Disease with Organ-                   ASCL2. Cell. Death Dis. 13, 884 (2022). https://doi.org/10.1038/
      oids. Cell. 165, 1586–1597 (2016). https://doi.org/10.1016/j.              s41419-022-05301-x
      cell.2016.05.082                                                     53.   P.Y. Hwang, J. Mathur, Y. Cao, J. Almeida, J. Ye, V. Morikis et
37.   M.A. Lancaster, J.A. Knoblich, Organogenesis in a dish: modeling           al., A Cdh3-β-catenin-laminin signaling axis in a subset of breast
      development and disease using organoid technologies. Science.              tumor leader cells control leader cell polarization and directional
      345, 1247125 (2014). https://doi.org/10.1126/science.1247125               collective migration. Dev. Cell. 58, 34–50e9 (2023). https://doi.
38.   G. Rossi, A. Manfrin, M.P. Lutolf, Progress and potential in               org/10.1016/j.devcel.2022.12.005
      organoid research. Nat. Rev. Genet. 19, 671–687 (2018). https://     54.   E. Henriet, H. Knutsdottir, E.M. Grasset, M. Dunworth, M.
      doi.org/10.1038/s41576-018-0051-9                                          Haynes, J.S. Bader et al., Triple negative breast tumors contain
39.   N.S. Corsini, J.A. Knoblich, Human organoids: New strate-                  heterogeneous cancer cells expressing distinct KRAS-dependent
      gies and methods for analyzing human development and dis-                  collective and disseminative invasion programs. Oncogene. 42,
      ease. Cell. 185, 2756–2769 (2022). https://doi.org/10.1016/j.              737–747 (2023). https://doi.org/10.1038/s41388-022-02586-2
      cell.2022.06.051                                                     55.   M.B. Henningsen, K. McWhan, V.S. Dam, M. Mele, K.R. Hau-
40.   Y.E. Bar-Ephraim, K. Kretzschmar, H. Clevers, Organoids                    erslev, N.C.S. Voss et al., Amplified Ca2 + dynamics and acceler-
      in immunological research. Nat. Rev. Immunol. 20, 279–293                  ated cell proliferation in breast cancer tissue during purinergic
      (2020). https://doi.org/10.1038/s41577-019-0248-y                          stimulation. Int. J. Cancer. 151, 1150–1165 (2022). https://doi.
41.   de L.F. Oliveira, D.M. Filho, B.L. Marques, G.F. Maciel, R.C.              org/10.1002/ijc.34147
      Parreira, do, J.R. Carmo Neto et al., Organoids as a novel           56.   C.J. Hanley, E. Henriet, O.K. Sirka, G.J. Thomas, A.J. Ewald,
      tool in modelling infectious diseases. Semin Cell Dev Biol.                Tumor-Resident Stromal cells promote breast Cancer Inva-
      2023;144:87–96. https://doi.org/10.1016/j.semcdb.2022.09.003               sion through Regulation of the basal phenotype. Mol. Cancer
42.   K.I. Votanopoulos, S. Forsythe, H. Sivakumar, A. Mazzocchi,                Res. 18, 1615–1622 (2020). https://doi.org/10.1158/1541-7786.
      J. Aleman, L. Miller et al., Model of patient-specific Immune-             MCR-20-0334
      enhanced organoids for immunotherapy screening: feasibil-            57.   S. Lee, T.V. Axelsen, A.P. Andersen, P. Vahl, S.F. Pedersen,
      ity study. Ann. Surg. Oncol. 27, 1956–1967 (2020). https://doi.            E. Boedtkjer, Disrupting Na+,HCO3–-cotransporter NBCn1
      org/10.1245/s10434-019-08143-8                                             (Slc4a7) delays murine breast cancer development. Oncogene.
43.   C. Lago, M. Gianesello, L. Santomaso, G. Leva, C. Ballabio, M.             35, 2112–2122 (2016). https://doi.org/10.1038/onc.2015.273
      Anderle et al., Medulloblastoma and high-grade glioma organoids      58.   F. Chen, J. Liu, R.M. Flight, K.J. Naughton, A. Lukyanchuk, A.R.
      for drug screening, lineage tracing, co-culture and in vivo assay.         Edgin et al., Cellular origins of EGFR-Driven Lung Cancer cells
      Nat. Protoc. 18, 2143–2180 (2023). https://doi.org/10.1038/                determine sensitivity to Therapy. Adv. Sci. 8, 2101999 (2021).
      s41596-023-00839-2                                                         https://doi.org/10.1002/advs.202101999
44.   F. Jacob, R.D. Salinas, D.Y. Zhang, P.T.T. Nguyen, J.G. Schnoll,     59.   J. Hai, H. Zhang, J. Zhou, Z. Wu, T. Chen, E. Papadopoulos et
      S.Z.H. Wong et al., A patient-derived glioblastoma organoid                al., Generation of genetically engineered mouse lung organoid
      model and biobank recapitulates Inter- and intra-tumoral hetero-           models for squamous cell lung cancers allows for the study of
      geneity. Cell. 180, 188–204e22 (2020). https://doi.org/10.1016/j.          combinatorial immunotherapy. Clin. Cancer Res. 26, 3431–3442
      cell.2019.11.036                                                           (2020). https://doi.org/10.1158/1078-0432.CCR-19-1627
45.   M.T. Meister, M.J.A. Groot Koerkamp, de T. Souza, W.B. Breu-         60.   A.F.M. Dost, A.L. Moye, M. Vedaie, L.M. Tran, E. Fung, D.
      nis, E. Frazer-Mendelewska, M. Brok et al., Mesenchymal tumor              Heinze et al., Organoids Model Transcriptional Hallmarks of
      organoid models recapitulate rhabdomyosarcoma subtypes.                    Oncogenic KRAS activation in lung epithelial progenitor cells.
13
A systematic review on the culture methods and applications of 3D tumoroids for cancer research and…
      Cell. Stem Cell. 27, 663–678e8 (2020). https://doi.org/10.1016/j.             tumorigenesis in mice. Cell. Rep. 41, 111482 (2022). https://doi.
      stem.2020.07.022                                                              org/10.1016/j.celrep.2022.111482
61.   P.P.G. Guimaraes, M. Tan, T. Tammela, K. Wu, A. Chung, M.               76.   K. Dixon, T. Brew, D. Farnell, T.D. Godwin, S. Cheung, C.
      Oberli et al., Potent in vivo lung cancer wnt signaling inhibi-               Chow et al., Modelling hereditary diffuse gastric cancer initia-
      tion via cyclodextrin-LGK974 inclusion complexes. J. Con-                     tion using transgenic mouse-derived gastric organoids and sin-
      trolled Release. 290, 75–87 (2018). https://doi.org/10.1016/j.                gle-cell sequencing. J. Pathol. 254, 254–264 (2021). https://doi.
      jconrel.2018.09.025                                                           org/10.1002/path.5675
62.   T. Sato, M. Morita, R. Tanaka, Y. Inoue, M. Nomura, Y. Sakamoto         77.   T. Seidlitz, Y.-T. Chen, H. Uhlemann, S. Schölch, S. Kochall,
      et al., Ex vivo model of non–small cell lung cancer using mouse               S.R. Merker et al., Mouse models of human gastric Cancer sub-
      lung epithelial cells. Oncol. Lett. 14, 6863–6868 (2017). https://            types with stomach-specific CreERT2-Mediated pathway altera-
      doi.org/10.3892/ol.2017.7098                                                  tions. Gastroenterology. 157, 1599–1614e2 (2019). https://doi.
63.   K.-S. Hsu, M. Adileh, M.L. Martin, V. Makarov, J. Chen, C.                    org/10.1053/j.gastro.2019.09.026
      Wu et al., Colorectal Cancer develops inherent radiosensitivity         78.   L.D. Nadauld, S. Garcia, G. Natsoulis, J.M. Bell, L. Miotke,
      that can be predicted using patient-derived Organoids. Cancer                 E.S. Hopmans et al., Metastatic tumor evolution and organoid
      Res. 82, 2298–2312 (2022). https://doi.org/10.1158/0008-5472.                 modeling implicate TGFBR2as a cancer driver in diffuse gastric
      CAN-21-4128                                                                   cancer. Genome Biol. 15, 428 (2014). https://doi.org/10.1186/
64.   J.W. Villanueva, L. Kwong, T. Han, S.A. Martinez, M.T. Shana-                 s13059-014-0428-9
      han, M. Kanke et al., Comprehensive microRNA analysis across            79.   J. Liu, P. Li, L. Wang, M. Li, Z. Ge, L. Noordam et al., Cancer-
      genome-edited colorectal cancer organoid models reveals miR-24                associated fibroblasts provide a stromal niche for liver cancer
      as a candidate regulator of cell survival. BMC Genom. 23, 792                 organoids that confers trophic effects and therapy resistance.
      (2022). https://doi.org/10.1186/s12864-022-09018-1                            Cell. Mol. Gastroenterol. Hepatol. 11, 407–431 (2021). https://
65.   A. Álvarez-Varela, L. Novellasdemunt, F.M. Barriga, X. Her-                   doi.org/10.1016/j.jcmgh.2020.09.003
      nando-Momblona, A. Cañellas-Socias, S. Cano-Crespo et al.,              80.   W. Cao, M. Li, J. Liu, S. Zhang, L. Noordam, M.M.A. Verstegen
      Mex3a marks drug-tolerant persister colorectal cancer cells that              et al., LGR5 marks targetable tumor-initiating cells in mouse liver
      mediate relapse after chemotherapy. Nat. Cancer. 3, 1052–1070                 cancer. Nat. Commun. 2020;11:1961. https://doi.org/10.1038/
      (2022). https://doi.org/10.1038/s43018-022-00402-0                            s41467-020-15846-0
66.   A. Fumagalli, K.C. Oost, L. Kester, J. Morgner, L. Bornes, L.           81.   A. Saborowski, K. Wolff, S. Spielberg, B. Beer, B. Hartleben, Z.
      Bruens et al., Plasticity of Lgr5-Negative Cancer cells drives                Erlangga et al., Murine liver organoids as a genetically flexible
      Metastasis in Colorectal Cancer. Cell. Stem Cell. 26, 569–578e7               system to Study Liver Cancer in vivo and in Vitro. Hepatol. Com-
      (2020). https://doi.org/10.1016/j.stem.2020.02.008                            mun. 3, 423–436 (2019). https://doi.org/10.1002/hep4.1312
67.   P. Cheung, J. Xiol, M.T. Dill, W.-C. Yuan, R. Panero, J. Roper et       82.   W. Cao, J. Liu, L. Wang, M. Li, M.M.A. Verstegen, Y. Yin et al.,
      al., Regenerative reprogramming of the intestinal stem cell state             Modeling liver cancer and therapy responsiveness using organ-
      via Hippo Signaling suppresses metastatic colorectal Cancer.                  oids derived from primary mouse liver tumors. Carcinogenesis.
      Cell. Stem Cell. 27, 590–604e9 (2020). https://doi.org/10.1016/j.             40, 145–154 (2019). https://doi.org/10.1093/carcin/bgy129
      stem.2020.07.003                                                        83.   M. Inglebert, M. Dettwiler, K. Hahn, A. Letko, C. Drogemuller, J.
68.   J.M. Chan, S. Zaidi, J.R. Love, J.L. Zhao, M. Setty, K.M. Wadosky             Doench et al., A living biobank of canine mammary tumor organ-
      et al., Lineage plasticity in prostate cancer depends on JAK/STAT             oids as a comparative model for human breast cancer. Sci. Rep.
      inflammatory signaling. Science. 377, 1180–1191 (2022). https://              12, 18051 (2022). https://doi.org/10.1038/s41598-022-21706-2
      doi.org/10.1126/science.abn0478                                         84.   C. Cocola, S. Molgora, E. Piscitelli, M.C. Veronesi, M. Greco,
69.   K.M. Wadosky, Y. Wang, X. Zhang, D.W. Goodrich, generation                    C. Bragato et al., FGF2 and EGF are required for Self-Renewal
      of tumor organoids from genetically engineered mouse models of                and Organoid formation of Canine Normal and Tumor breast
      prostate cancer. J. Vis. Exp. 2019. https://doi.org/10.3791/59710             stem cells. J. Cell. Biochem. 118, 570–584 (2017). https://doi.
70.   M. Blattner, D. Liu, B.D. Robinson, D. Huang, A. Poliakov, D.                 org/10.1002/jcb.25737
      Gao et al., SPOP mutation drives prostate tumorigenesis in vivo         85.   Y. Shiota, M. Elbadawy, K. Suzuki, R. Tsunedomi, H. Nagano, Y.
      through coordinate regulation of PI3K/mTOR and AR signal-                     Ishihara, Derivation of a new model of lung adenocarcinoma using
      ing. Cancer Cell. 31, 436–451 (2017). https://doi.org/10.1016/j.              canine lung cancer organoids for translational research in pulmo-
      ccell.2017.02.004                                                             nary medicine et al., (eds.), Biomedicine & Pharmacotherapy
71.   E. Dardenne, H. Beltran, M. Benelli, K. Gayvert, A. Berger, L.                2023;165:115079. https://doi.org/10.1016/j.biopha.2023.115079
      Puca et al., N-Myc induces an EZH2-mediated transcriptional             86.   L. Chandra, D.C. Borcherding, D. Kingsbury, T. Atherly, Y.M.
      program driving neuroendocrine prostate Cancer. Cancer Cell.                  Ambrosini, A. Bourgois-Mochel et al., Derivation of adult
      30, 563–577 (2016). https://doi.org/10.1016/j.ccell.2016.09.005               canine intestinal organoids for translational research in gastro-
72.   S. Agarwal, P.G. Hynes, H.S. Tillman, R. Lake, W.G. Abou-                     enterology. BMC Biol. 17, 33 (2019). https://doi.org/10.1186/
      Kheir, L. Fang et al., Identification of different classes of Luminal         s12915-019-0652-6
      Progenitor cells within prostate tumors. Cell. Rep. 13, 2147–2158       87.   T. Usui, M. Sakurai, S. Nishikawa, K. Umata, Y. Nemoto, T.
      (2015). https://doi.org/10.1016/j.celrep.2015.10.077                          Haraguchi et al., Establishment of a dog primary prostate can-
73.   W.R. Karthaus, P.J. Iaquinta, J. Drost, A. Gracanin, van R. Boxtel,           cer organoid using the urine cancer stem cells. Cancer Sci. 108,
      J. Wongvipat et al., Identification of multipotent luminal progeni-           2383–2392 (2017). https://doi.org/10.1111/cas.13418
      tor cells in human prostate organoid cultures. Cell. 159, 163–175       88.   A. Abugomaa, M. Elbadawy, M. Yamanaka, Y. Goto, K. Hayashi,
      (2014). https://doi.org/10.1016/j.cell.2014.08.017                            T. Mori et al., Establishment of 2.5D organoid culture model
74.   C.W. Chua, M. Shibata, M. Lei, R. Toivanen, L.J. Barlow, S.K.                 using 3D bladder cancer organoid culture. Sci. Rep. 10, 9393
      Bergren et al., Single luminal epithelial progenitors can generate            (2020). https://doi.org/10.1038/s41598-020-66229-w
      prostate organoids in culture. Nat. Cell. Biol. 16, 951–954 (2014).     89.   M. Elbadawy, T. Usui, T. Mori, R. Tsunedomi, S. Hazama, R.
      https://doi.org/10.1038/ncb3047                                               Nabeta et al., Establishment of a novel experimental model
75.   Z. Lu, A. Zhong, H. Liu, M. Zhang, X. Chen, X. Pan et al., Dis-               for muscle-invasive bladder cancer using a dog bladder cancer
      secting the genetic and microenvironmental factors of gastric                 organoid culture. Cancer Sci. 110, 2806–2821 (2019). https://doi.
                                                                                    org/10.1111/cas.14118
                                                                                                                                          13
                                                                                                                                    J. Kalla et al.
90. S. Scheemaeker, M. Inglebert, S. Daminet, M. Dettwiler, A.            105. A.P. Bartlett, R.M. Harman, J.R. Weiss, Van de G.R. Walle,
     Letko, C. Drögemüller et al., Organoids of patient-derived medul-         Establishment and characterization of equine mammary organ-
     lary thyroid carcinoma: the first milestone towards a new in vitro        oids using a method translatable to other non-traditional model
     model in dogs. Vet. Comp. Oncol. 21, 111–122 (2023). https://             species. Development. 149, dev200412 (2022). https://doi.
     doi.org/10.1111/vco.12872                                                 org/10.1242/dev.200412
91. J. Jankovic, M. Dettwiler, M.G. Fernández, E. Tièche, K. Hahn,        106. H. Li, Y. Wang, M. Zhang, H. Wang, A. Cui, J. Zhao et al., Estab-
     S. April-Monn et al., Validation of immunohistochemistry for              lishment of porcine and monkey colonic organoids for drug tox-
     canine proteins involved in thyroid iodine uptake and their               icity study. Cell. Regen. 10, 32 (2021). https://doi.org/10.1186/
     expression in Canine Follicular cell thyroid carcinomas (FTCs)            s13619-021-00094-4
     and FTC-Derived Organoids. Vet. Pathol. 58, 1172–1180 (2021).        107. V. Veninga, E.E. Voest, Tumor organoids: opportunities and chal-
     https://doi.org/10.1177/03009858211018813                                 lenges to guide precision medicine. Cancer Cell. 39, 1190–1201
92. M.M. Callesen, S.S. Árnadóttir, I. Lyskjær, M.W. Ørntoft, S.               (2021). https://doi.org/10.1016/j.ccell.2021.07.020
     Høyer, F. Dagnæs-Hansen et al., A genetically inducible porcine      108. K.P. Guillen, M. Fujita, A.J. Butterfield, S.D. Scherer, M.H.
     model of intestinal cancer. Mol. Oncol. 11, 1616–1629 (2017).             Bailey, Z. Chu et al., A human breast cancer-derived xenograft
     https://doi.org/10.1002/1878-0261.12136                                   and organoid platform for drug discovery and precision oncol-
93. W. Hill, D.R. Caswell, C. Swanton, Capturing cancer evolution              ogy. Nat. Cancer. 3, 232–250 (2022). https://doi.org/10.1038/
     using genetically engineered mouse models (GEMMs). Trends                 s43018-022-00337-6
     Cell Biol. 31, 1007–1018 (2021). https://doi.org/10.1016/j.          109. J.M. Rosenbluth, R.C.J. Schackmann, G.K. Gray, L.M. Selfors,
     tcb.2021.07.003                                                           C.M.-C. Li, M. Boedicker et al., Organoid cultures from nor-
94. S. Zhang, S. Iyer, H. Ran, I. Dolgalev, S. Gu, W. Wei et al.,              mal and cancer-prone human breast tissues preserve complex
     Genetically defined, Syngeneic Organoid platform for develop-             epithelial lineages. Nat. Commun. 11, 1711 (2020). https://doi.
     ing combination therapies for ovarian Cancer. Cancer Discov. 11,          org/10.1038/s41467-020-15548-7
     362–383 (2021). https://doi.org/10.1158/2159-8290.CD-20-0455         110. N. Sachs, de J. Ligt, O. Kopper, E. Gogola, G. Bounova, F. Wee-
95. K. Lõhmussaar, O. Kopper, J. Korving, H. Begthel, C.P.H. Vreuls,           ber et al., A living biobank of breast Cancer Organoids captures
     van J.H. Es et al., Assessing the origin of high-grade serous             Disease Heterogeneity. Cell. 172, 373–386e10 (2018). https://doi.
     ovarian cancer using CRISPR-modification of mouse organ-                  org/10.1016/j.cell.2017.11.010
     oids. Nat. Commun. 11, 2660 (2020). https://doi.org/10.1038/         111. M. Kim, H. Mun, C.O. Sung, E.J. Cho, H.-J. Jeon, S.-M. Chun
     s41467-020-16432-0                                                        et al., Patient-derived lung cancer organoids as in vitro cancer
96. R.R.J. Low, K.Y. Fung, H. Gao, A. Preaudet, L.F. Dagley, J.                models for therapeutic screening. Nat. Commun. 10, 3991 (2019).
     Yousef et al., S100 family proteins are linked to organoid mor-           https://doi.org/10.1038/s41467-019-11867-6
     phology and EMT in pancreatic cancer. Cell. Death Differ. 30,        112. C. Pauli, B.D. Hopkins, D. Prandi, R. Shaw, T. Fedrizzi, A.
     1155–1165 (2023). https://doi.org/10.1038/s41418-023-01126-z              Sboner et al., Personalized in Vitro and in vivo Cancer models
97. K. Cheaito, H.F. Bahmad, O. Hadadeh, H. Msheik, A. Monzer,                 to Guide Precision Medicine. Cancer Discov. 7, 462–477 (2017).
     F. Ballout et al., Establishment and characterization of pros-            https://doi.org/10.1158/2159-8290.CD-16-1154
     tate organoids from treatment–naïve patients with prostate           113. K. Toshimitsu, A. Takano, M. Fujii, K. Togasaki, M. Matano, S.
     cancer. Oncol. Lett. 23, 1–16 (2022). https://doi.org/10.3892/            Takahashi et al., Organoid screening reveals epigenetic vulnera-
     ol.2021.13124                                                             bilities in human colorectal cancer. Nat. Chem. Biol. 18, 605–614
98. S. Pamarthy, H.E. Sabaawy, Patient derived organoids in pros-              (2022). https://doi.org/10.1038/s41589-022-00984-x
     tate cancer: improving therapeutic efficacy in precision medi-       114. M. Fujii, M. Shimokawa, S. Date, A. Takano, M. Matano, K.
     cine. Mol. Cancer. 20, 125 (2021). https://doi.org/10.1186/               Nanki et al., A colorectal tumor Organoid Library demon-
     s12943-021-01426-3                                                        strates progressive loss of Niche factor requirements during
99. I.W. Mak, N. Evaniew, M. Ghert, Lost in translation: animal mod-           Tumorigenesis. Cell. Stem Cell. 18, 827–838 (2016). https://doi.
     els and clinical trials in cancer treatment. Am. J. Transl Res. 6,        org/10.1016/j.stem.2016.04.003
     114–118 (2014)                                                       115. R. Servant, M. Garioni, T. Vlajnic, M. Blind, H. Pueschel, D.C.
100. C.H.C. Leenaars, C. Kouwenaar, F.R. Stafleu, A. Bleich, M.                Müller et al., Prostate cancer patient-derived organoids: detailed
     Ritskes-Hoitinga, De R.B.M. Vries et al., Animal to human trans-          outcome from a prospective cohort of 81 clinical specimens. J.
     lation: a systematic scoping review of reported concordance rates.        Pathol. 254, 543–555 (2021). https://doi.org/10.1002/path.5698
     J. Translational Med. 17, 223 (2019). https://doi.org/10.1186/       116. H.H.N. Yan, H.C. Siu, S. Law, S.L. Ho, S.S.K. Yue, W.Y. Tsui et
     s12967-019-1976-2                                                         al., A Comprehensive Human gastric Cancer Organoid Biobank
101. M. Kawasaki, T. Goyama, Y. Tachibana, I. Nagao, Y.M. Ambro-               captures Tumor Subtype Heterogeneity and enables therapeutic
     sini, Farm and Companion Animal Organoid models in Transla-               screening. Cell. Stem Cell. 23, 882–897e11 (2018). https://doi.
     tional Research: a powerful Tool to Bridge the Gap between mice           org/10.1016/j.stem.2018.09.016
     and humans. Front. Med. Technol. 2022;4                              117. K. Nanki, K. Toshimitsu, A. Takano, M. Fujii, M. Shimokawa,
102. M. Elbadawy, K. Fujisaka, H. Yamamoto, R. Tsunedomi, H.                   Y. Ohta et al., Divergent routes toward wnt and R-spondin Niche
     Nagano, H. Ayame et al., Establishment of an experimental model           Independency during Human gastric carcinogenesis. Cell. 174,
     of normal dog bladder organoid using a three-dimensional culture          856–869e17 (2018). https://doi.org/10.1016/j.cell.2018.07.027
     method. Biomed. Pharmacother. 151, 113105 (2022). https://doi.       118. G. Vlachogiannis, S. Hedayat, A. Vatsiou, Y. Jamin, J. Fernández-
     org/10.1016/j.biopha.2022.113105                                          Mateos, K. Khan et al., Patient-derived organoids model treat-
103. M. Elbadawy, Y. Sato, T. Mori, Y. Goto, K. Hayashi, M. Yamanaka           ment response of metastatic gastrointestinal cancers. Science.
     et al., Anti-tumor effect of trametinib in bladder cancer organoid        359, 920–926 (2018). https://doi.org/10.1126/science.aao2774
     and the underlying mechanism. Cancer Biol. Ther. 22, 357–371         119. L. Broutier, G. Mastrogiovanni, M.M. Verstegen, H.E. Francies,
     (2021). https://doi.org/10.1080/15384047.2021.1919004                     L.M. Gavarró, C.R. Bradshaw et al., Human primary liver cancer-
104. A. Abugomaa, M. Elbadawy, Y. Ishihara, H. Yamamoto, M.                    derived organoid cultures for disease modeling and drug screen-
     Kaneda, H. Yamawaki et al., Anti-cancer activity of Chaga mush-           ing. Nat. Med. 23, 1424–1435 (2017). https://doi.org/10.1038/
     room (Inonotus Obliquus) against dog bladder cancer organoids.            nm.4438
     Front. Pharmacol. 2023;14
13
A systematic review on the culture methods and applications of 3D tumoroids for cancer research and…
120. H.S. Seol, J.H. Oh, E. Choi, S. Kim, H. Kim, E.J. Nam, Preclini-           Nat. Cell. Biol. 21, 1041–1051 (2019). https://doi.org/10.1038/
     cal investigation of patient-derived cervical cancer organoids for         s41556-019-0360-z
     precision medicine. J. Gynecol. Oncol. 34, e35 (2022). https://       135. Y. Nanki, T. Chiyoda, A. Hirasawa, A. Ookubo, M. Itoh, M.
     doi.org/10.3802/jgo.2023.34.e35                                            Ueno et al., Patient-derived ovarian cancer organoids capture the
121. K. Lõhmussaar, R. Oka, J. Espejo Valle-Inclan, M.H.H. Smits, H.            genomic profiles of primary tumours applicable for drug sensitiv-
     Wardak, J. Korving et al., Patient-derived organoids model cervi-          ity and resistance testing. Sci. Rep. 10, 12581 (2020). https://doi.
     cal tissue dynamics and viral oncogenesis in cervical cancer. Cell.        org/10.1038/s41598-020-69488-9
     Stem Cell. 28, 1380–1396e6 (2021). https://doi.org/10.1016/j.         136. N. Maenhoudt, C. Defraye, M. Boretto, Z. Jan, R. Heremans, B.
     stem.2021.03.012                                                           Boeckx et al., Developing Organoids from Ovarian Cancer as
122. T.A. Karakasheva, J.T. Gabre, U.M. Sachdeva, R. Cruz-Acuña,                Experimental and Preclinical models. Stem Cell. Rep. 14, 717–
     E.W. Lin, M. DeMarshall et al., Patient-derived organoids as a             729 (2020). https://doi.org/10.1016/j.stemcr.2020.03.004
     platform for modeling a patient’s response to chemoradiotherapy       137. A. Linkous, H.A. Fine, Generating patient-derived gliomas
     in esophageal cancer. Sci. Rep. 11, 21304 (2021). https://doi.             within cerebral organoids. STAR. Protocols. 1, 100008 (2020).
     org/10.1038/s41598-021-00706-8                                             https://doi.org/10.1016/j.xpro.2019.100008
123. T. Kijima, H. Nakagawa, M. Shimonosono, P.M. Chandramou-              138. A. Linkous, D. Balamatsias, M. Snuderl, L. Edwards, K. Miyagu-
     leeswaran, T. Hara, V. Sahu et al., Three-Dimensional Organoids            chi, T. Milner et al., Modeling patient-derived glioblastoma with
     reveal Therapy Resistance of esophageal and oropharyngeal                  cerebral organoids. Cell. Rep. 26, 3203–3211e5 (2019). https://
     squamous cell carcinoma cells. Cell. Mol. Gastroenterol. Hepa-             doi.org/10.1016/j.celrep.2019.02.063
     tol. 7, 73–91 (2019). https://doi.org/10.1016/j.jcmgh.2018.09.003     139. C.G. Hubert, M. Rivera, L.C. Spangler, Q. Wu, S.C. Mack,
124. X. Li, H.E. Francies, M. Secrier, J. Perner, A. Miremadi, N. Gale-         B.C. Prager et al., A three-Dimensional Organoid Culture Sys-
     ano-Dalmau et al., Organoid cultures recapitulate esophageal               tem Derived from Human Glioblastomas recapitulates the
     adenocarcinoma heterogeneity providing a model for clonality               hypoxic gradients and Cancer stem cell heterogeneity of tumors
     studies and precision therapeutics. Nat. Commun. 9, 2983 (2018).           found in vivo. Cancer Res. 76, 2465–2477 (2016). https://doi.
     https://doi.org/10.1038/s41467-018-05190-9                                 org/10.1158/0008-5472.CAN-15-2402
125. H. Yang, Q. Liang, J. Zhang, J. Liu, H. Wei, H. Chen et al., Estab-   140. M. Perréard, R. Florent, J. Divoux, J.-M. Grellard, J. Lequesne,
     lishment of papillary thyroid cancer organoid lines from clinical          M. Briand et al., ORGAVADS: establishment of tumor organ-
     specimens. Front. Endocrinol. 2023;14                                      oids from head and neck squamous cell carcinoma to assess their
126. M. Romitti, A. Tourneur, de B. Faria da Fonseca, G. Doumont, P.            response to innovative therapies. BMC Cancer. 23, 223 (2023).
     Gillotay, X.-H. Liao et al., Transplantable human thyroid organ-           https://doi.org/10.1186/s12885-023-10692-x
     oids generated from embryonic stem cells to rescue hypothyroid-       141. E. Driehuis, S. Kolders, S. Spelier, K. Lõhmussaar, S.M. Wil-
     ism. Nat. Commun. 13, 7057 (2022). https://doi.org/10.1038/                lems, L.A. Devriese et al., Oral mucosal organoids as a poten-
     s41467-022-34776-7                                                         tial platform for Personalized Cancer Therapy. Cancer Discov. 9,
127. D. Chen, Y. Tan, Z. Li, W. Li, L. Yu, W. Chen et al., Organoid             852–871 (2019). https://doi.org/10.1158/2159-8290.CD-18-1522
     cultures derived from patients with papillary thyroid Cancer. J.      142. N. Tanaka, A.A. Osman, Y. Takahashi, A. Lindemann, A.A. Patel,
     Clin. Endocrinol. Metab. 106, 1410–1426 (2021). https://doi.               M. Zhao et al., Head and neck cancer organoids established
     org/10.1210/clinem/dgab020                                                 by modification of the CTOS method can be used to predict in
128. M. Minoli, T. Cantore, D. Hanhart, M. Kiener, T. Fedrizzi, La              vivo drug sensitivity. Oral Oncol. 87, 49–57 (2018). https://doi.
     F. Manna et al., Bladder cancer organoids as a functional sys-             org/10.1016/j.oraloncology.2018.10.018
     tem to model different disease stages and therapy response.           143. A.R. Mazzocchi, S.A.P. Rajan, K.I. Votanopoulos, A.R. Hall, A.
     Nat. Commun. 14, 2214 (2023). https://doi.org/10.1038/                     Skardal, In vitro patient-derived 3D mesothelioma tumor organ-
     s41467-023-37696-2                                                         oids facilitate patient-centric therapeutic screening. Sci. Rep. 8,
129. S.H. Lee, W. Hu, J.T. Matulay, M.V. Silva, T.B. Owczarek, K.               2886 (2018). https://doi.org/10.1038/s41598-018-21200-8
     Kim et al., Tumor evolution and drug response in patient-derived      144. Z. Li, H. Xu, L. Yu, J. Wang, Q. Meng, H. Mei et al., Patient-
     Organoid models of bladder Cancer. Cell. 173, 515–528e17                   derived renal cell carcinoma organoids for personalized cancer
     (2018). https://doi.org/10.1016/j.cell.2018.03.017                         therapy. Clin. Translational Med. 12, e970 (2022). https://doi.
130. H. Tiriac, P. Belleau, D.D. Engle, D. Plenker, A. Deschênes,               org/10.1002/ctm2.970
     T.D.D. Somerville et al., Organoid profiling identifies common        145. H.A. Bolck, C. Corrò, A. Kahraman, von A. Teichman, N.C.
     responders to chemotherapy in pancreatic cancer. Cancer Dis-               Toussaint, J. Kuipers et al., Tracing Clonal Dynamics reveals
     cov. 8, 1112–1129 (2018). https://doi.org/10.1158/2159-8290.               that two- and three-dimensional patient-derived cell models cap-
     CD-18-0349                                                                 ture Tumor Heterogeneity of Clear Cell Renal Cell Carcinoma.
131. L. Huang, A. Holtzinger, I. Jagan, M. BeGora, I. Lohse, N. Ngai et         Eur. Urol. Focus. 7, 152–162 (2021). https://doi.org/10.1016/j.
     al., Ductal pancreatic cancer modeling and drug screening using            euf.2019.06.009
     human pluripotent stem cell and patient-derived tumor organ-          146. L. Grassi, R. Alfonsi, F. Francescangeli, M. Signore, De M.L.
     oids. Nat. Med. 21, 1364–1371 (2015). https://doi.org/10.1038/             Angelis, A. Addario et al., Organoids as a new model for improv-
     nm.3973                                                                    ing regenerative medicine and cancer personalized therapy
132. S.F. Boj, C.-I. Hwang, L.A. Baker, I.I.C. Chio, D.D. Engle,                in renal diseases. Cell. Death Dis. 10, 201 (2019). https://doi.
     V. Corbo et al., Organoid models of Human and Mouse Duc-                   org/10.1038/s41419-019-1453-0
     tal Pancreatic Cancer. Cell. 160, 324–338 (2015). https://doi.        147. S.D. Forsythe, R.A. Erali, P. Laney, H. Sivakumar, W. Li, A. Skar-
     org/10.1016/j.cell.2014.12.021                                             dal et al., Application of immune enhanced organoids in modeling
133. A. Katcher, B. Yueh, K. Ozler, A. Nizam, A. Kredentser, C.                 personalized Merkel cell carcinoma research. Sci. Rep. 12, 13865
     Chung et al., Establishing patient-derived organoids from human            (2022). https://doi.org/10.1038/s41598-022-17921-6
     endometrial cancer and normal endometrium. Front. Endocrinol.         148. R.A. DeStefanis, J.D. Kratz, A.M. Olson, A. Sunil, A.K.
     2023;14                                                                    DeZeeuw, A.A. Gillette et al., Impact of baseline culture condi-
134. M. Boretto, N. Maenhoudt, X. Luo, A. Hennes, B. Boeckx, B. Bui             tions of cancer organoids when determining therapeutic response
     et al., Patient-derived organoids from endometrial disease cap-            and tumor heterogeneity. Sci. Rep. 12, 5205 (2022). https://doi.
     ture clinical heterogeneity and are amenable to drug screening.            org/10.1038/s41598-022-08937-z
                                                                                                                                       13
                                                                                                                                     J. Kalla et al.
149. V. Narasimhan, J.A. Wright, M. Churchill, T. Wang, R. Rosati,             interpatient drug response heterogeneity. JCI Insight. 4, e121490
     T.R.M. Lannagan et al., Medium-throughput drug screening of               (2019). https://doi.org/10.1172/jci.insight.121490
     patient-derived Organoids from Colorectal Peritoneal metastases      164. M. Dhainaut, S.A. Rose, G. Akturk, A. Wroblewska, S.R. Nielsen,
     to Direct Personalized Therapy. Clin. Cancer Res. 26, 3662–3670           E.S. Park et al., Spatial CRISPR genomics identifies regulators of
     (2020). https://doi.org/10.1158/1078-0432.CCR-20-0073                     the tumor microenvironment. Cell. 185, 1223–1239e20 (2022).
150. S.N. Ooft, F. Weeber, K.K. Dijkstra, C.M. McLean, S. Kaing,               https://doi.org/10.1016/j.cell.2022.02.015
     van E. Werkhoven et al., Patient-derived organoids can predict       165. L. Brady, M. Kriner, I. Coleman, C. Morrissey, M. Roudier, L.D.
     response to chemotherapy in metastatic colorectal cancer patients.        True et al., Inter- and intra-tumor heterogeneity of metastatic
     Sci. Transl Med. 11, eaay2574 (2019). https://doi.org/10.1126/            prostate cancer determined by digital spatial gene expression pro-
     scitranslmed.aay2574                                                      filing. Nat. Commun. 12, 1426 (2021). https://doi.org/10.1038/
151. Y. Mao, W. Wang, J. Yang, X. Zhou, Y. Lu, J. Gao et al., Drug             s41467-021-21615-4
     repurposing screening and mechanism analysis based on human          166. W. Senkowski, L. Gall-Mas, M.M. Falco, Y. Li, K. Lavikka, M.C.
     colorectal cancer organoids. Protein Cell. pwad038 (2023).                Kriegbaum et al., A platform for efficient establishment and drug-
     https://doi.org/10.1093/procel/pwad038                                    response profiling of high-grade serous ovarian cancer organoids.
152. S. Hahn, B.J. Oh, H. Kim, I.W. Han, S.H. Shin, G. Kim et al.,             Dev. Cell. 58, 1106–1121e7 (2023). https://doi.org/10.1016/j.
     Anti-cancer effects of metformin in a 3D co-culture model of              devcel.2023.04.012
     pancreatic ductal adenocarcinoma. Am. J. Cancer Res. 13, 1806–       167. S. Mo, P. Tang, W. Luo, L. Zhang, Y. Li, X. Hu et al., Patient-
     1825 (2023)                                                               derived Organoids from Colorectal Cancer with Paired Liver
153. C.K. Hirt, T.H. Booij, L. Grob, P. Simmler, N.C. Toussaint,               Metastasis reveal Tumor heterogeneity and predict response to
     D. Keller et al., Drug screening and genome editing in human              Chemotherapy. Adv. Sci. (Weinh). 9, e2204097 (2022). https://
     pancreatic cancer organoids identifies drug-gene interactions             doi.org/10.1002/advs.202204097
     and candidates for off-label treatment. Cell. Genom. 2, 100095       168. J.E. Grossman, L. Muthuswamy, L. Huang, D. Akshinthala, S.
     (2022). https://doi.org/10.1016/j.xgen.2022.100095                        Perea, R.S. Gonzalez et al., Organoid Sensitivity correlates with
154. T.T. Seppälä, J.W. Zimmerman, R. Suri, H. Zlomke, G.D. Ivey, A.           therapeutic response in patients with pancreatic Cancer. Clin.
     Szabolcs et al., Precision Medicine in Pancreatic Cancer: patient-        Cancer Res. 28, 708–718 (2022). https://doi.org/10.1158/1078-
     derived Organoid Pharmacotyping is a predictive biomarker of              0432.CCR-20-4116
     clinical treatment response. Clin. Cancer Res. 28, 3296–3307         169. C.-C. Chen, H.-W. Li, Y.-L. Wang, C.-C. Lee, Y.-C. Shen, C.-Y.
     (2022). https://doi.org/10.1158/1078-0432.CCR-21-4165                     Hsieh et al., Patient-derived tumor organoids as a platform of pre-
155. K. Geevimaan, J.-Y. Guo, C.-N. Shen, J.-K. Jiang, C.S.J. Fann,            cision treatment for malignant brain tumors. Sci. Rep. 12, 16399
     M.-J. Hwang et al., Patient-derived Organoid serves as a plat-            (2022). https://doi.org/10.1038/s41598-022-20487-y
     form for personalized chemotherapy in Advanced Colorectal            170. P. Horak, C. Heining, S. Kreutzfeldt, B. Hutter, A. Mock, J.
     Cancer patients. Front. Oncol. 12, 883437 (2022). https://doi.            Hüllein et al., Comprehensive genomic and transcriptomic
     org/10.3389/fonc.2022.883437                                              analysis for guiding therapeutic decisions in patients with rare
156. A. Hennig, F. Baenke, A. Klimova, S. Drukewitz, B. Jahnke, S.             cancers. Cancer Discov. 11, 2780–2795 (2021). https://doi.
     Brückmann et al., Detecting drug resistance in pancreatic cancer          org/10.1158/2159-8290.CD-21-0126
     organoids guides optimized chemotherapy treatment. J. Pathol.        171. de E. Heus, S.F.A. Duijts, van der J.M. Zwan, E. Kapiteijn, E.J.M.
     257, 607–619 (2022). https://doi.org/10.1002/path.5906                    van Nieveen, van C.M.L. Herpen et al., The gap between rare
157. K. Harada, N. Sakamoto, S. Ukai, Y. Yamamoto, Q.T. Pham, D.               and common cancers still exists: results from a population-based
     Taniyama et al., Establishment of oxaliplatin-resistant gastric           study in the Netherlands. Eur. J. Cancer. 167, 103–111 (2022).
     cancer organoids: importance of myoferlin in the acquisition of           https://doi.org/10.1016/j.ejca.2022.03.001
     oxaliplatin resistance. Gastric Cancer. 24, 1264–1277 (2021).        172. M.-A. Meier, S. Nuciforo, M. Coto-Llerena, J. Gallon, M.S. Mat-
     https://doi.org/10.1007/s10120-021-01206-4                                ter, C. Ercan et al., Patient-derived tumor organoids for personal-
158. S. Ukai, R. Honma, N. Sakamoto, Y. Yamamoto, Q.T. Pham, K.                ized medicine in a patient with rare hepatocellular carcinoma with
     Harada et al., Molecular biological analysis of 5-FU-resistant            neuroendocrine differentiation: a case report. Commun. Med. 2,
     gastric cancer organoids; KHDRBS3 contributes to the attain-              1–8 (2022). https://doi.org/10.1038/s43856-022-00150-3
     ment of features of cancer stem cell. Oncogene. 39, 7265–7278        173. G. Liu, X. Xiao, Y. Xia, W. Huang, W. Chen, J. Xu et al., Organ-
     (2020). https://doi.org/10.1038/s41388-020-01492-9                        oids from Mucinous Appendiceal Adenocarcinomas as High-
159. S.L. Boos, L.P. Loevenich, S. Vosberg, T. Engleitner, R. Öllinger,        Fidelity models for Individual Therapy. Front. Med. (Lausanne).
     J. Kumbrink et al., Disease modeling on Tumor Organoids impli-            9, 829033 (2022). https://doi.org/10.3389/fmed.2022.829033
     cates AURKA as a therapeutic target in Liver metastatic colorec-     174. S.-Y. Kim, S.-M. Kim, S. Lim, J.Y. Lee, S.-J. Choi, S.-D. Yang et
     tal Cancer. Cell. Mol. Gastroenterol. Hepatol. 13, 517–540                al., Modeling clinical responses to targeted therapies by patient-
     (2021). https://doi.org/10.1016/j.jcmgh.2021.10.008                       derived organoids of Advanced Lung Adenocarcinoma. Clin.
160. R. Vegliante, I. Pastushenko, C. Blanpain, Deciphering functional         Cancer Res. 27, 4397–4409 (2021). https://doi.org/10.1158/1078-
     tumor states at single-cell resolution. EMBO J. 41, e109221               0432.CCR-20-5026
     (2022). https://doi.org/10.15252/embj.2021109221                     175. K. Ganesh, C. Wu, K.P. O’Rourke, B.C. Szeglin, Y. Zheng, C.-
161. L. González-Silva, L. Quevedo, I. Varela, Tumor Functional Het-           E.G. Sauvé et al., A rectal cancer organoid platform to study indi-
     erogeneity unraveled by scRNA-seq technologies. Trends Can-               vidual responses to chemoradiation. Nat. Med. 25, 1607–1614
     cer. 6, 13–19 (2020). https://doi.org/10.1016/j.trecan.2019.11.010        (2019). https://doi.org/10.1038/s41591-019-0584-2
162. de C.J. Witte, J. Espejo Valle-Inclan, N. Hami, K. Lõhmussaar,       176. Y. Yao, X. Xu, L. Yang, J. Zhu, J. Wan, L. Shen et al., Patient-
     O. Kopper, C.P.H. Vreuls et al., Patient-derived ovarian Cancer           derived Organoids Predict Chemoradiation responses of locally
     Organoids Mimic Clinical response and exhibit Heterogeneous               advanced rectal Cancer. Cell. Stem Cell. 26, 17–26e6 (2020).
     Inter- and intrapatient drug responses. Cell. Rep. 31, 107762             https://doi.org/10.1016/j.stem.2019.10.010
     (2020). https://doi.org/10.1016/j.celrep.2020.107762                 177. R. Millen, De W.W.B. Kort, M. Koomen, van G.J.F. Son, R.
163. L. Li, H. Knutsdottir, K. Hui, M.J. Weiss, J. He, B. Philosophe et        Gobits, B. Penning de Vries et al., Patient-derived head and neck
     al., Human primary liver cancer organoids reveal intratumor and           cancer organoids allow treatment stratification and serve as a tool
13
A systematic review on the culture methods and applications of 3D tumoroids for cancer research and…
     for biomarker validation and identification. Med. 4, 290–310e12           interactions. Physiol. Rep. 2, e12147 (2014). https://doi.
     (2023). https://doi.org/10.1016/j.medj.2023.04.003                        org/10.14814/phy2.12147
178. S.J. Sundar, S. Shakya, A. Barnett, L.C. Wallace, H. Jeon, A.        194. J.M. McFarland, B.R. Paolella, A. Warren, K. Geiger-Schuller, T.
     Sloan et al., Three-dimensional organoid culture unveils resis-           Shibue, M. Rothberg et al., Multiplexed single-cell transcriptional
     tance to clinical therapies in adult and pediatric glioblastoma.          response profiling to define cancer vulnerabilities and therapeutic
     Transl Oncol. 15, 101251 (2021). https://doi.org/10.1016/j.               mechanism of action. Nat. Commun. 11, 4296 (2020). https://doi.
     tranon.2021.101251                                                        org/10.1038/s41467-020-17440-w
179. R.-X. Huang, P.-K. Zhou, DNA damage response signaling path-         195. B. Chen, C.R. Scurrah, E.T. McKinley, A.J. Simmons, M.A.
     ways and targets for radiotherapy sensitization in cancer. Sig            Ramirez-Solano, X. Zhu et al., Differential pre-malignant pro-
     Transduct. Target. Ther. 5, 1–27 (2020). https://doi.org/10.1038/         grams and microenvironment chart distinct paths to malignancy
     s41392-020-0150-x                                                         in human colorectal polyps. Cell. 184, 6262–6280e26 (2021).
180. de K.E. Visser, J.A. Joyce, The evolving tumor microenviron-              https://doi.org/10.1016/j.cell.2021.11.031
     ment: from cancer initiation to metastatic outgrowth. Cancer Cell.   196. A. Ootani, S. Toda, K. Fujimoto, H. Sugihara, An air–liquid inter-
     41, 374–403 (2023). https://doi.org/10.1016/j.ccell.2023.02.016           face promotes the differentiation of gastric surface mucous cells
181. Z. Xiao, Z. Dai, J.W. Locasale, Metabolic landscape of the tumor          (GSM06) in culture. Biochem. Biophys. Res. Commun. 271,
     microenvironment at single cell resolution. Nat. Commun. 10,              741–746 (2000). https://doi.org/10.1006/bbrc.2000.2673
     3763 (2019). https://doi.org/10.1038/s41467-019-11738-0              197. S.L. Giandomenico, S.B. Mierau, G.M. Gibbons, L.M.D.
182. D.C. Hinshaw, L.A. Shevde, The Tumor Microenvironment                     Wenger, L. Masullo, T. Sit et al., Cerebral organoids at the air-
     innately modulates Cancer Progression. Cancer Res. 79, 4557–              liquid interface generate diverse nerve tracts with functional out-
     4566 (2019). https://doi.org/10.1158/0008-5472.CAN-18-3962                put. Nat. Neurosci. 22, 669–679 (2019). https://doi.org/10.1038/
183. J.T. Neal, X. Li, J. Zhu, V. Giangarra, C.L. Grzeskowiak, J. Ju           s41593-019-0350-2
     et al., Organoid modeling of the Tumor Immune Microenviron-          198. X. Li, A. Ootani, C. Kuo, An air-liquid Interface Culture Sys-
     ment. Cell. 175, 1972–1988e16 (2018). https://doi.org/10.1016/j.          tem for 3D Organoid Culture of Diverse primary gastrointesti-
     cell.2018.11.021                                                          nal tissues. Methods Mol. Biol. 1422, 33–40 (2016). https://doi.
184. E.M. Langer, B.L. Allen-Petersen, S.M. King, N.D. Kendsersky,             org/10.1007/978-1-4939-3603-8_4
     M.A. Turnidge, G.M. Kuziel et al., Modeling Tumor Phenotypes         199. H. Kobayashi, A. Enomoto, S.L. Woods, A.D. Burt, M. Taka-
     in Vitro with three-Dimensional Bioprinting. Cell. Rep. 26, 608–          hashi, D.L. Worthley, Cancer-associated fibroblasts in gastroin-
     623e6 (2019). https://doi.org/10.1016/j.celrep.2018.12.090                testinal cancer. Nat. Rev. Gastroenterol. Hepatol. 16, 282–295
185. P. Datta, M. Dey, Z. Ataie, D. Unutmaz, I.T. Ozbolat, 3D bioprint-        (2019). https://doi.org/10.1038/s41575-019-0115-0
     ing for reconstituting the cancer microenvironment. Npj Precis       200. E. Sahai, I. Astsaturov, E. Cukierman, D.G. DeNardo, M. Ege-
     Onc. 4, 1–13 (2020). https://doi.org/10.1038/s41698-020-0121-2            blad, R.M. Evans et al., A framework for advancing our under-
186. V.S. Shirure, Y. Bi, M.B. Curtis, A. Lezia, M.M. Goedegebuure,            standing of cancer-associated fibroblasts. Nat. Rev. Cancer. 20,
     S.P. Goedegebuure et al., Tumor-on-a-chip platform to investi-            174–186 (2020). https://doi.org/10.1038/s41568-019-0238-1
     gate progression and drug sensitivity in cell lines and patient-     201. E.M. Zeisberg, S. Potenta, L. Xie, M. Zeisberg, R. Kalluri, Dis-
     derived organoids. Lab. Chip. 18, 3687–3702 (2018). https://doi.          covery of endothelial to Mesenchymal Transition as a source for
     org/10.1039/c8lc00596f                                                    Carcinoma-Associated fibroblasts. Cancer Res. 67, 10123–10128
187. S. Schuth, Le S. Blanc, T.G. Krieger, J. Jabs, M. Schenk, N.A.            (2007). https://doi.org/10.1158/0008-5472.CAN-07-3127
     Giese et al., Patient-specific modeling of stroma-mediated chemo-    202. H. Dang, T.J. Harryvan, C.-Y. Liao, E.H.J. Danen, V.N.L.N.
     resistance of pancreatic cancer using a three-dimensional organ-          Spalburg, S.M. Kielbasa et al., Cancer-Associated fibroblasts are
     oid-fibroblast co-culture system. J. Experimental Clin. Cancer            key determinants of Cancer Cell Invasion in the Earliest Stage of
     Res. 41, 312 (2022). https://doi.org/10.1186/s13046-022-02519-7           Colorectal Cancer. Cell. Mol. Gastroenterol. Hepatol. 16, 107–
188. V.S. Atanasova, C. Cardona, de J. Hejret, V. Tiefenbacher, A.             131 (2023). https://doi.org/10.1016/j.jcmgh.2023.04.004
     Mair, T. Tran, Mimicking Tumor Cell Heterogeneity of Colorec-        203. E. Strating, M.P. Verhagen, E. Wensink, E. Dünnebach, L. Wijler,
     tal Cancer in a patient-derived Organoid-Fibroblast Model. Cell.          I. Aranguren et al., Co-cultures of colon cancer cells and can-
     Mol. Gastroenterol. Hepatol. 15, 1391–1419 (2023). https://doi.           cer-associated fibroblasts recapitulate the aggressive features of
     org/10.1016/j.jcmgh.2023.02.014                                           mesenchymal-like colon cancer. Front. Immunol. 2023;14
189. J. Chakrabarti, V. Koh, J.B.Y. So, W.P. Yong, Y. Zavros, A Pre-      204. M.H. Mosa, B.E. Michels, C. Menche, A.M. Nicolas, T. Darvishi,
     clinical Human-Derived Autologous Gastric Cancer Organoid/                F.R. Greten et al., A wnt-Induced phenotypic switch in Cancer-
     Immune Cell Co-culture Model to predict the efficacy of targeted          Associated fibroblasts inhibits EMT in Colorectal Cancer. Cancer
     therapies. J. Vis. Exp. (2021). https://doi.org/10.3791/61443             Res. 80, 5569–5582 (2020). https://doi.org/10.1158/0008-5472.
190. X. Qin, J. Sufi, P. Vlckova, P. Kyriakidou, S.E. Acton, V.S.W.            CAN-20-0263
     Li et al., Cell-type specific signalling networks in Heterocellu-    205. L. Shaashua, A. Ben-Shmuel, M. Pevsner-Fischer, G. Friedman,
     lar Organoids. Nat. Methods. 17, 335–342 (2020). https://doi.             O. Levi-Galibov, S. Nandakumar et al., BRCA mutational sta-
     org/10.1038/s41592-020-0737-8                                             tus shapes the stromal microenvironment of pancreatic cancer
191. S. Jiang, T. Deng, H. Cheng, W. Liu, D. Shi, J. Yuan et al., Mac-         linking clusterin expression in cancer associated fibroblasts with
     rophage-organoid co-culture model for identifying treatment               HSF1 signaling. Nat. Commun. 13, 6513 (2022). https://doi.
     strategies against macrophage-related gemcitabine resistance.             org/10.1038/s41467-022-34081-3
     J. Experimental Clin. Cancer Res. 42, 199 (2023). https://doi.       206. J.F. Dekkers, M. Alieva, A. Cleven, F. Keramati, A.K.L. Weze-
     org/10.1186/s13046-023-02756-4                                            naar, van E.J. Vliet et al., Uncovering the mode of action of engi-
192. H. Fang, Y. Huang, Y. Luo, J. Tang, M. Yu, Y. Zhang et al., SIRT1         neered T cells in patient cancer organoids. Nat. Biotechnol. 41,
     induces the accumulation of TAMs at colorectal cancer tumor               60–69 (2023). https://doi.org/10.1038/s41587-022-01397-w
     sites via the CXCR4/CXCL12 axis. Cell. Immunol. 371, 104458          207. T.E. Schnalzger, de M.H. Groot, C. Zhang, M.H. Mosa, B.E.
     (2022). https://doi.org/10.1016/j.cellimm.2021.104458                     Michels, J. Röder et al., 3D model for CAR-mediated cytotoxic-
193. Y.-G. Zhang, S. Wu, Y. Xia, J. Sun, Salmonella-infected crypt-            ity using patient-derived colorectal cancer organoids. EMBO J.
     derived intestinal organoid culture system for host-bacterial             38, e100928 (2019). https://doi.org/10.15252/embj.2018100928
                                                                                                                                     13
                                                                                                                                        J. Kalla et al.
208. I.S. Chan, H. Knútsdóttir, G. Ramakrishnan, V. Padmanaban,                   Adv. Mater. Technol. 8, 2201942 (2023). https://doi.org/10.1002/
     M. Warrier, J.C. Ramirez et al., Cancer cells educate natural                admt.202201942
     killer cells to a metastasis-promoting cell state. J. Cell Biol. 219,   224. D. Nieto, G. Jiménez, L. Moroni, E. López-Ruiz, P. Gálvez‐
     e202001134 (2020). https://doi.org/10.1083/jcb.202001134                     Martín, J.A. Marchal, Biofabrication approaches and regulatory
209. L.E. Wagar, B. Sworder, M.S. Khodadoust, M.M. Davis, A.A.                    framework of metastatic tumor‐on‐a‐chip models for precision
     Alizadeh, Follicular lymphoma organoids for investigating the                oncology. Med. Res. Rev. 42, 1978–2001 (2022). https://doi.
     Tumor Microenvironment. Blood. 134, 2799 (2019). https://doi.                org/10.1002/med.21914
     org/10.1182/blood-2019-131192                                           225. A.M.K. Law, L. Rodriguez de la Fuente, T.J. Grundy, G. Fang, F.
210. A. Rogoz, B.S. Reis, R.A. Karssemeijer, D. Mucida, A 3-D                     Valdes-Mora, D. Gallego-Ortega, Advancements in 3D cell Cul-
     enteroid-based model to study T-cell and epithelial cell inter-              ture systems for personalizing Anti-cancer therapies. Front. Oncol.
     action. J. Immunol. Methods. 421, 89–95 (2015). https://doi.                 11, 782766 (2021). https://doi.org/10.3389/fonc.2021.782766
     org/10.1016/j.jim.2015.03.014                                           226. S.N. Bhatia, D.E. Ingber, Microfluidic organs-on-chips. Nat. Bio-
211. C. Wan, M.P. Keany, H. Dong, L.F. Al-Alem, U.M. Pandya,                      technol. 32, 760–772 (2014). https://doi.org/10.1038/nbt.2989
     S. Lazo et al., Enhanced efficacy of simultaneous PD-1 and              227. A. Sontheimer-Phelps, B.A. Hassell, D.E. Ingber, Modelling can-
     PD-L1 Immune Checkpoint Blockade in High-Grade Serous                        cer in microfluidic human organs-on-chips. Nat. Rev. Cancer. 19,
     Ovarian Cancer. Cancer Res. 81, 158–173 (2021). https://doi.                 65–81 (2019). https://doi.org/10.1038/s41568-018-0104-6
     org/10.1158/0008-5472.CAN-20-1674                                       228. D. Pinho, D. Santos, A. Vila, S. Carvalho, Establishment
212. K. Hezaveh, R.S. Shinde, A. Klötgen, M.J. Halaby, S. Lamorte,                of Colorectal Cancer organoids in Microfluidic-based sys-
     M.T. Ciudad et al., Tryptophan-derived microbial metabolites                 tem. Micromachines. 12, 497 (2021). https://doi.org/10.3390/
     activate the aryl hydrocarbon receptor in tumor-associated mac-              mi12050497
     rophages to suppress anti-tumor immunity. Immunity. 55, 324–            229. P. Mehta, Z. Rahman, ten P. Dijke, P.E. Boukany, Microfluid-
     340e8 (2022). https://doi.org/10.1016/j.immuni.2022.01.006                   ics meets 3D cancer cell migration. Trends Cancer. 8, 683–697
213. S. Chen, W. Cui, Z. Chi, Q. Xiao, T. Hu, Q. Ye et al., Tumor-                (2022). https://doi.org/10.1016/j.trecan.2022.03.006
     associated macrophages are shaped by intratumoral high potas-           230. D.J. Jung, T.H. Shin, M. Kim, C.O. Sung, S.J. Jang, G.S. Jeong,
     sium via Kir2.1. Cell Metabol. 34, 1843–1859e11 (2022). https://             A one-stop microfluidic-based lung cancer organoid culture
     doi.org/10.1016/j.cmet.2022.08.016                                           platform for testing drug sensitivity. Lab. Chip. 19, 2854–2865
214. Y.-C. Wang, X. Wang, J. Yu, F. Ma, Z. Li, Y. Zhou et al., Targeting          (2019). https://doi.org/10.1039/C9LC00496C
     monoamine oxidase A-regulated tumor-associated macrophage               231. B. Schuster, M. Junkin, S.S. Kashaf, I. Romero-Calvo, K.
     polarization for cancer immunotherapy. Nat. Commun. 12, 3530                 Kirby, J. Matthews et al., Automated microfluidic platform for
     (2021). https://doi.org/10.1038/s41467-021-23164-2                           dynamic and combinatorial drug screening of tumor organ-
215. J. Sufi, X. Qin, F.C. Rodriguez, Y.J. Bu, P. Vlckova, M.R. Zapa-             oids. Nat. Commun. 11, 5271 (2020). https://doi.org/10.1038/
     tero et al., Multiplexed single-cell analysis of organoid signal-            s41467-020-19058-4
     ing networks. Nat. Protoc. 16, 4897–4918 (2021). https://doi.           232. F.L. Lai Benjamin, X. Lu Rick, Y. Hu, H.L. Davenport, W. Dou,
     org/10.1038/s41596-021-00603-4                                               E.Y. Wang et al., Recapitulating pancreatic tumor microenviron-
216. R. Augustine, S.N. Kalva, R. Ahmad, A.A. Zahid, S. Hasan, A.                 ment through synergistic use of patient organoids and organ-on-a-
     Nayeem et al., 3D bioprinted cancer models: revolutionizing                  chip vasculature. Adv. Funct. Mater. 30, 2000545 (2020). https://
     personalized cancer therapy. Transl Oncol. 14, 101015 (2021).                doi.org/10.1002/adfm.202000545
     https://doi.org/10.1016/j.tranon.2021.101015                            233. M.R. Haque, C.R. Wessel, D.D. Leary, C. Wang, A. Bhushan, F.
217. A. Mazzocchi, S. Soker, A. Skardal, 3D bioprinting for high-                 Bishehsari, Patient-derived pancreatic cancer-on-a-chip recapitu-
     throughput screening: drug screening, disease modeling, and pre-             lates the tumor microenvironment. Microsyst. Nanoeng. 8, 1–13
     cision medicine applications. Appl. Phys. Rev. 6, 011302 (2019).             (2022). https://doi.org/10.1038/s41378-022-00370-6
     https://doi.org/10.1063/1.5056188                                       234. M. Geyer, D. Schreyer, L.-M. Gaul, S. Pfeffer, C. Pilarsky, K.
218. J. Kim, J. Jang, D.-W. Cho, Recapitulating the Cancer Micro-                 Queiroz, A microfluidic-based PDAC organoid system reveals
     environment using Bioprinting Technology for Precision Medi-                 the impact of hypoxia in response to treatment. Cell. Death Dis-
     cine. Micromachines. 12, 1122 (2021). https://doi.org/10.3390/               cov. 9, 1–8 (2023). https://doi.org/10.1038/s41420-023-01334-z
     mi12091122                                                              235. C.-P. Day, G. Merlino, Van T. Dyke, Preclinical mouse Cancer
219. C. Calandrini, J. Drost, Normal and tumor-derived organoids as               models: a maze of opportunities and challenges. Cell. 163, 39–53
     a drug screening platform for tumor-specific drug vulnerabili-               (2015). https://doi.org/10.1016/j.cell.2015.08.068
     ties. STAR. Protoc. 3, 101079 (2022). https://doi.org/10.1016/j.        236. E. Wang, K. Xiang, Y. Zhang, X.-F. Wang, Patient-derived organ-
     xpro.2021.101079                                                             oids (PDOs) and PDO-derived xenografts (PDOXs): new oppor-
220. H. Chen, Z. Wu, Z. Gong, Y. Xia, J. Li, L. Du et al., Acoustic               tunities in establishing faithful pre-clinical cancer models. J.
     bioprinting of patient-derived Organoids for Predicting Cancer               Natl. Cancer Cent. 2, 263–276 (2022). https://doi.org/10.1016/j.
     therapy responses. Adv. Healthc. Mater. 11, 2102784 (2022).                  jncc.2022.10.001
     https://doi.org/10.1002/adhm.202102784                                  237. Y. Maru, N. Tanaka, Y. Tatsumi, Y. Nakamura, M. Itami, Y. Hippo,
221. Y. Choi, H. Lee, M. Ann, M. Song, J. Rheey, J. Jang, 3D bio-                 Kras activation in endometrial organoids drives cellular transfor-
     printed vascularized lung cancer organoid models with underlying             mation and epithelial-mesenchymal transition. Oncogenesis. 10,
     disease capable of more precise drug evaluation. Biofabrication.             1–12 (2021). https://doi.org/10.1038/s41389-021-00337-8
     15, 034104 (2023). https://doi.org/10.1088/1758-5090/acd95f             238. S. Karkampouna, La F. Manna, A. Benjak, M. Kiener, De M.
222. E. Maloney, C. Clark, H. Sivakumar, K. Yoo, J. Aleman, S.A.P.                Menna, E. Zoni et al., Patient-derived xenografts and organoids
     Rajan et al., Immersion bioprinting of Tumor organoids in Multi-             model therapy response in prostate cancer. Nat. Commun. 12,
     well plates for increasing Chemotherapy Screening Throughput.                1117 (2021). https://doi.org/10.1038/s41467-021-21300-6
     Micromachines (Basel). 11, 208 (2020). https://doi.org/10.3390/         239. K. Liu, N. Jing, D. Wang, P. Xu, J. Wang, X. Chen et al., A novel
     mi11020208                                                                   mouse model for liver metastasis of prostate cancer reveals
223. Z. Gong, Y. Mao, L. Huang, X. Tang, L. Zhang, Y. Xu et al.,                  dynamic tumour-immune cell communication. Cell Prolif. 54,
     Acoustic Printing of patient-derived Organoids that preserve                 e13056 (2021). https://doi.org/10.1111/cpr.13056
     Tumor Microenvironment for Personalized Drug Screening.
13
A systematic review on the culture methods and applications of 3D tumoroids for cancer research and…
240. K. Miyabayashi, L.A. Baker, A. Deschênes, B. Traub, G. Caligi-        254. A.L. Bredenoord, H. Clevers, J.A. Knoblich, Human tissues in a
     uri, D. Plenker et al., Intraductal transplantation models of human        dish: the research and ethical implications of organoid technol-
     pancreatic ductal adenocarcinoma reveal progressive transition             ogy. Science. 355, eaaf9414 (2017). https://doi.org/10.1126/sci-
     of molecular subtypes. Cancer Discov. 10, 1566–1589 (2020).                ence.aaf9414
     https://doi.org/10.1158/2159-8290.CD-20-0133                          255. S.N. Ooft, F. Weeber, L. Schipper, K.K. Dijkstra, C.M. McLean,
241. S. Lee, T.R. Mendoza, D.N. Burner, M.T. Muldong, C.C.N. Wu,                S. Kaing et al., Prospective experimental treatment of colorectal
     C. Arreola-Villanueva et al., Novel dormancy mechanism of                  cancer patients based on organoid drug responses. ESMO Open.
     Castration Resistance in Bone metastatic prostate Cancer Organ-            6, 100103 (2021). https://doi.org/10.1016/j.esmoop.2021.100103
     oids. Int. J. Mol. Sci. 23, 3203 (2022). https://doi.org/10.3390/     256. E.A. Aisenbrey, W.L. Murphy, Synthetic alternatives to Matri-
     ijms23063203                                                               gel. Nat. Rev. Mater. 5, 539–551 (2020). https://doi.org/10.1038/
242. H. Dillekås, M.S. Rogers, O. Straume, Are 90% of deaths from               s41578-020-0199-8
     cancer caused by metastases? Cancer Med. 8, 5574 (2019).              257. M.T. Kozlowski, C.J. Crook, H.T. Ku, Towards organoid culture
     https://doi.org/10.1002/cam4.2474                                          without Matrigel. Commun. Biol. 4, 1387 (2021). https://doi.
243. D. Lin, L. Shen, M. Luo, K. Zhang, J. Li, Q. Yang et al., Cir-             org/10.1038/s42003-021-02910-8
     culating tumor cells: biology and clinical significance. Sig          258. S. Kim, S. Min, Y.S. Choi, S.-H. Jo, J.H. Jung, K. Han et al.,
     Transduct. Target. Ther. 6, 1–24 (2021). https://doi.org/10.1038/          Tissue extracellular matrix hydrogels as alternatives to Matrigel
     s41392-021-00817-8                                                         for culturing gastrointestinal organoids. Nat. Commun. 13, 1692
244. De M.L. Angelis, F. Francescangeli, C. Nicolazzo, M. Signore, A.           (2022). https://doi.org/10.1038/s41467-022-29279-4
     Giuliani, L. Colace et al., An organoid model of colorectal circu-    259. K.K. Dijkstra, K. Monkhorst, L.J. Schipper, K.J. Hartemink,
     lating tumor cells with stem cell features, hybrid EMT state and           E.F. Smit, S. Kaing et al., Challenges in establishing pure Lung
     distinctive therapy response profile. J. Experimental Clin. Cancer         Cancer Organoids Limit their utility for Personalized Medi-
     Res. 41, 86 (2022). https://doi.org/10.1186/s13046-022-02263-y             cine. Cell. Rep. 31, 107588 (2020). https://doi.org/10.1016/j.
245. De M.L. Angelis, F. Francescangeli, C. Nicolazzo, E. Xhelili, La           celrep.2020.107588
     F. Torre, L. Colace et al., An Orthotopic patient-derived xeno-       260. M.C. Skala, D.A. Deming, J.D. Kratz, Technologies to assess
     graft (PDX) model allows the analysis of Metastasis-Associated             drug response and heterogeneity in patient-derived Cancer
     features in Colorectal Cancer. Front. Oncol. 12, 869485 (2022).            Organoids. Annu. Rev. Biomed. Eng. 24, 157–177 (2022). https://
     https://doi.org/10.3389/fonc.2022.869485                                   doi.org/10.1146/annurev-bioeng-110220-123503
246. E.M. Grasset, M. Dunworth, G. Sharma, M. Loth, J. Tandurella,         261. O.H. Usman, L. Zhang, G. Xie, H.M. Kocher, C. Hwang, Y.J.
     A. Cimino-Mathews et al., Triple-negative breast cancer metasta-           Wang et al., Genomic heterogeneity in pancreatic cancer organ-
     sis involves complex epithelial-mesenchymal transition dynam-              oids and its stability with culture. Npj Genom Med. 7, 1–10
     ics and requires vimentin. Sci. Transl Med. 14, eabn7571 (2022).           (2022). https://doi.org/10.1038/s41525-022-00342-9
     https://doi.org/10.1126/scitranslmed.abn7571                          262. Y. Du, X. Li, Q. Niu, X. Mo, M. Qui, T. Ma et al., Development
247. A. Golebiewska, A.-C. Hau, A. Oudin, D. Stieber, Y.A. Yabo, V.             of a miniaturized 3D organoid culture platform for ultra-high-
     Baus et al., Patient-derived organoids and orthotopic xenografts           throughput screening. J. Mol. Cell. Biol. 12, 630–643 (2020).
     of primary and recurrent gliomas represent relevant patient ava-           https://doi.org/10.1093/jmcb/mjaa036
     tars for precision oncology. Acta Neuropathol. 140, 919–949           263. S. Hou, H. Tiriac, B.P. Sridharan, L. Scampavia, F. Madoux,
     (2020). https://doi.org/10.1007/s00401-020-02226-7                         J. Seldin et al., Advanced Development of primary pancreatic
248. H.F. Berg, M.E. Hjelmeland, H. Lien, H. Espedal, T. Fonnes,                organoid tumor models for high-throughput phenotypic drug
     A. Srivastava et al., Patient-derived organoids reflect the                screening. SLAS Discovery. 23, 574–584 (2018). https://doi.
     genetic profile of endometrial tumors and predict patient prog-            org/10.1177/2472555218766842
     nosis. Commun. Med. 1, 1–14 (2021). https://doi.org/10.1038/          264. P.J. Tebon, B. Wang, A.L. Markowitz, A. Davarifar, B.L. Tsai, P.
     s43856-021-00019-x                                                         Krawczuk et al., Drug screening at single-organoid resolution via
249. P. Tan, M. Wang, A. Zhong, Y. Wang, J. Du, J. Wang et al.,                 bioprinting and interferometry. Nat. Commun. 14, 3168 (2023).
     SRT1720 inhibits the growth of bladder cancer in organoids                 https://doi.org/10.1038/s41467-023-38832-8
     and murine models through the SIRT1-HIF axis. Oncogene. 40,           265. N. Phan, J.J. Hong, B. Tofig, M. Mapua, D. Elashoff, N.A.
     6081–6092 (2021). https://doi.org/10.1038/s41388-021-01999-9               Moatamed et al., A simple high-throughput approach identifies
250. J. Chen, L. Cao, Z. Li, Y. Li, SIRT1 promotes GLUT1 expres-                actionable drug sensitivities in patient-derived tumor organ-
     sion and bladder cancer progression via regulation of glucose              oids. Commun. Biol. 2, 1–11 (2019). https://doi.org/10.1038/
     uptake. Hum. Cell. 32, 193–201 (2019). https://doi.org/10.1007/            s42003-019-0305-x
     s13577-019-00237-5                                                    266. L.M. Ortiz Jordan, V.F. Vega, J. Shumate, A. Peles, J. Zeiger, L.
251. Q. Hu, G. Wang, J. Peng, G. Qian, W. Jiang, C. Xie, Y. Xiao,               Scampavia et al., Protocol for high throughput 3D drug screen-
     X. Wang, 2017. Knockdown of SIRT1 Suppresses Bladder Can-                  ing of patient derived melanoma and renal cell carcinoma.
     cer Cell Proliferation and Migration and Induces Cell Cycle                SLAS Discovery. 100141 (2024). https://doi.org/10.1016/j.
     Arrest and Antioxidant Response through FOXO3a-Medi-                       slasd.2024.01.002
     ated Pathways. Biomed Res Int 2017, 3781904. https://doi.             267. D.B. Gunasekara, M. DiSalvo, Y. Wang, D.L. Nguyen, M.I.
     org/10.1155/2017/3781904                                                   Reed, J. Speer et al., Development of arrayed colonic organoids
252. J.O.R. Hernandez, X. Wang, M. Vazquez-Segoviano, M. Lopez-                 for screening of secretagogues associated with enterotoxins.
     Marfil, M.F. Sobral-Reyes, A. Moran-Horowich et al., A tissue-             Anal. Chem. 90, 1941–1950 (2018). https://doi.org/10.1021/acs.
     bioengineering strategy for modeling rare human kidney diseases            analchem.7b04032
     in vivo. Nat. Commun. 12, 6496 (2021). https://doi.org/10.1038/       268. X. Li, G. Fu, L. Zhang, R. Guan, P. Tang, J. Zhang et al., Assay
     s41467-021-26596-y                                                         establishment and validation of a high-throughput organoid-
253. C. Gatzweiler, J. Ridinger, S. Herter, X.F. Gerloff, D. ElHarouni,         based drug screening platform. Stem Cell Res. Ther. 13, 219
     Y. Berker et al., Functional therapeutic target validation using           (2022). https://doi.org/10.1186/s13287-022-02902-3
     Pediatric zebrafish xenograft models. Cancers (Basel). 14, 849        269. S.M. Czerniecki, N.M. Cruz, J.L. Harder, R. Menon, J. Annis,
     (2022). https://doi.org/10.3390/cancers14030849                            E.A. Otto et al., High-throughput automation enhances kidney
                                                                                organoid differentiation from human pluripotent stem cells and
                                                                                                                                     13
                                                                                                                                       J. Kalla et al.
     enables multidimensional phenotypic screening. Cell. Stem Cell.            Numerical Simulation. Adv. Biol. (Weinh). 5, e2000024 (2021).
     22, 929 (2018). https://doi.org/10.1016/j.stem.2018.04.022                 https://doi.org/10.1002/adbi.202000024
270. I. Lukonin, M. Zinner, P. Liberali, Organoids in image-based phe-     283. D. Mathur, E. Barnett, H.I. Scher, J.B. Xavier, Optimizing the
     notypic chemical screens. Exp. Mol. Med. 53, 1495–1502 (2021).             future: how mathematical models inform treatment sched-
     https://doi.org/10.1038/s12276-021-00641-8                                 ules for cancer. Trends Cancer. 8, 506–516 (2022). https://doi.
271. F.L. Lampart, D. Iber, N. Doumpas, Organoids in high-through-              org/10.1016/j.trecan.2022.02.005
     put and high-content screenings. Front. Chem. Eng. 2023;5             284. A. Zuchowska, S. Skorupska, Multi-organ-on-chip approach in
272. P. Mukashyaka, P. Kumar, D.J. Mellert, S. Nicholas, J. Noor-               cancer research. Organs-on-a-Chip. 4, 100014 (2022). https://doi.
     bakhsh, M. Brugiolo et al., High-throughput deconvolution of               org/10.1016/j.ooc.2021.100014
     3D organoid dynamics at cellular resolution for cancer pharma-        285. A. Skardal, S.V. Murphy, M. Devarasetty, I. Mead, H.-W. Kang,
     cology with Cellos. Nat. Commun. 14, 8406 (2023). https://doi.             Y.-J. Seol et al., Multi-tissue interactions in an integrated three-
     org/10.1038/s41467-023-44162-6                                             tissue organ-on-a-chip platform. Sci. Rep. 7, 8837 (2017). https://
273. Z. Zhao, X. Chen, A.M. Dowbaj, A. Sljukic, K. Bratlie, L. Lin et           doi.org/10.1038/s41598-017-08879-x
     al., Organoids Nat. Rev. Methods Primers. 2, 1–21 (2022). https://    286. X. Xie, X. Li, W. Song, Tumor organoid biobank-new platform
     doi.org/10.1038/s43586-022-00174-y                                         for medical research. Sci. Rep. 13, 1819 (2023). https://doi.
274. M.J. Kratochvil, A.J. Seymour, T.L. Li, S.P. Paşca, C.J. Kuo,              org/10.1038/s41598-023-29065-2
     S.C. Heilshorn, Engineered materials for organoid systems.            287. M.A. Foo, M. You, S.L. Chan, G. Sethi, G.K. Bonney, W.-P. Yong
     Nat. Rev. Mater. 4, 606–622 (2019). https://doi.org/10.1038/               et al., Clinical translation of patient-derived tumour organoids-
     s41578-019-0129-9                                                          bottlenecks and strategies. Biomark. Res. 10, 10 (2022). https://
275. M.F.B. Jamaluddin, A. Ghosh, A. Ingle, R. Mohammed, A.                     doi.org/10.1186/s40364-022-00356-6
     Ali, M. Bahrami et al., Bovine and human endometrium-                 288. A. Fuhr, A. Kurtz, C. Hiepen, S. Müller, Organoids as Miniature
     derived hydrogels support organoid culture from healthy and                Twins—challenges for comparability and need for data stan-
     cancerous tissues. Proceedings of the National Academy of                  dardization and Access. Organoids. 1, 28–36 (2022). https://doi.
     Sciences.     2022;119:e2208040119.        https://doi.org/10.1073/        org/10.3390/organoids1010003
     pnas.2208040119                                                       289. Patient-Derived Models Repository (PDMR) n.d. https://pdmr.
276. S. Price, S. Bhosle, E. Gonçalves, X. Li, D.P. McClurg, S. Bar-            cancer.gov/ (accessed August 1, 2023)
     thorpe et al., A suspension technique for efficient large-scale       290. Human Cancer Models Initiative - NCI, 2022. https://www.
     cancer organoid culturing and perturbation screens. Sci. Rep. 12,          cancer.gov/ccg/research/functional-genomics/hcmi          (accessed
     5571 (2022). https://doi.org/10.1038/s41598-022-09508-y                    August 1, 2023)
277. J.M. Unagolla, A.C. Jayasuriya, Recent advances in organoid           291. Home| HCA|Organoid, n.d. https://hca-organoid.eu/ (accessed
     engineering: a comprehensive review. Appl. Mater. Today. 29,               August 1, 2023)
     101582 (2022). https://doi.org/10.1016/j.apmt.2022.101582             292. C. Bock, M. Boutros, J.G. Camp, L. Clarke, H. Clevers, J.A.
278. Y. Cui, R. Xiao, Y. Zhou, J. Liu, Y. Wang, X. Yang et al., Estab-          Knoblich et al., Organoid cell. Atlas Nat. Biotechnol. 39, 13–17
     lishment of organoid models based on a nested array chip for               (2021). https://doi.org/10.1038/s41587-020-00762-x
     fast and reproducible drug testing in colorectal cancer therapy.      293. Q. Ma, H. Tao, Q. Li, Z. Zhai, X. Zhang, Z. Lin et al., Organ-
     Bio-Des Manuf. 5, 674–686 (2022). https://doi.org/10.1007/                 oidDB: a comprehensive organoid database for the multi-perspec-
     s42242-022-00206-2                                                         tive exploration of bulk and single-cell transcriptomic profiles of
279. S. Jiang, H. Zhao, W. Zhang, J. Wang, Y. Liu, Y. Cao et al., An            organoids. Nucleic Acids Res. 51, D1086–D1093 (2023). https://
     Automated Organoid platform with inter-organoid homogene-                  doi.org/10.1093/nar/gkac942
     ity and inter-patient heterogeneity. CR Med. 2020;1. https://doi.     294. R.J. Porter, G.I. Murray, M.H. McLean, Current concepts in
     org/10.1016/j.xcrm.2020.100161                                             tumour-derived organoids. Br. J. Cancer. 123, 1209–1218 (2020).
280. N. Brandenberg, S. Hoehnel, F. Kuttler, K. Homicsko, C. Ceroni,            https://doi.org/10.1038/s41416-020-0993-5
     T. Ringel et al., High-throughput automated organoid culture via      295. R.R.-K. Sen. Paul, All Info - S.5002–117th Congress (2021–
     stem-cell aggregation in microcavity arrays. Nat. Biomed. Eng. 4,          2022): FDA Modernization Act 2.0 [WWW Document]. (2022).
     863–874 (2020). https://doi.org/10.1038/s41551-020-0565-2                  https://www.congress.gov/bill/117th-congress/senate-bill/5002/
281. A. Vasciaveo, J.M. Arriaga, de F.N. Almeida, M. Zou, E.F. Jr               all-info (accessed 5.2.24)
     Douglass, F. Picech et al., OncoLoop: A Network-based Preci-
     sion Cancer Medicine Framework. Cancer Discov. 13, 386–409            Publisher’s Note Springer Nature remains neutral with regard to juris-
     (2023). https://doi.org/10.1158/2159-8290.CD-22-0342                  dictional claims in published maps and institutional affiliations.
282. F. Zheng, Y. Xiao, H. Liu, Y. Fan, M. Dao, Patient-specific Organ-
     oid and Organ-on-a-Chip: 3D cell-culture meets 3D Printing and
13