Stratification of colorectal cancer into subgroups with different response to therapy was initially guided by descriptive associations between specific biomarkers and treatment outcome. Recently, preclinical models based on propagatable patient-derived tumor samples have yielded an improved understanding of disease biology, which has facilitated the functional validation of correlative information and the discovery of novel response determinants, therapeutic targets, and mechanisms of tumor adaptation and drug resistance. We review the contribution of patient-derived models to advancing colorectal cancer characterization, discuss their influence on clinical decision-making, and highlight emerging challenges in the interpretation and clinical transferability of results obtainable with such approaches.

Significance:

Association studies in patients with colorectal cancer have led to the identification of response biomarkers, some of which have been implemented as companion diagnostics for therapeutic decisions. By enabling biological investigation in a clinically relevant experimental context, patient-derived colorectal cancer models have proved useful to examine the causal role of such biomarkers in dictating drug sensitivity and are providing fresh knowledge on new actionable targets, dynamics of tumor evolution and adaptation, and mechanisms of drug resistance.

Colorectal cancer is the third most frequent cause of cancer-related death in both the United States and Europe (1, 2). Around 25% of individuals harbor metastatic disease at the time of diagnosis, and approximately 50% of patients will develop metastases later (3). Although 5-year survival rates experienced by patients with metastatic colorectal cancer (mCRC) remain low (14%; ref. 1), therapeutic options developed in the past two decades have prolonged the median overall survival (OS) from 12 to 30 months (4). This survival advantage can be attributed to improved surgical techniques and the use of more effective systemic therapies. At least partially, more informed treatment decisions based on molecular response predictors have also helped increase life expectancy, but biomarker recognition has been slow and often inconclusive due to the difficulty of substantiating correlative observations in patients with functional investigation in clinically relevant model systems. Similarly, although genomic datasets have offered an instructive compendium of the genes that are frequently altered in colorectal cancer (5), the question of whether the aberrant protein products of such genes represent effective therapeutic targets remains hard to address in the absence of adequate translational tools.

The availability of large collections of patient-derived tumor samples that can be propagated in mice (xenografts) and in three-dimensional cultures (organoids) has spearheaded attempts to afford biomarker–response associations with mechanistic annotation and has facilitated studies aimed to model cancer progression and acquisition of drug resistance (6, 7). Herein, we provide an overview of current therapies and related biomarkers, as implemented in patients with mCRC, and discuss how patient-derived xenografts and organoids have been deployed to go beyond correlative descriptions and to illuminate fundamental biological and clinical aspects of colorectal cancer, including drug repurposing efforts that have rapidly moved to the clinical space. Further, we consider the practical implications and the limitations of using such models in terms of clinical applicability and predictivity.

The standard-of-care treatment for patients with mCRC includes cytotoxic agents and biological targeted compounds, which are administered cumulatively based on the empirical observation that multiagent therapeutic cocktails are more effective than monotherapies (8, 9). Although some patients receive important clinical benefit from these regimens, responses are typically limited to a fraction of individuals. The polarized distribution of responsive and nonresponsive patients likely derives from the genomic and functional heterogeneity of mCRC tumors, which display patient-to-patient molecular differences that influence treatment outcome. Although the application of “omics” technologies has been instrumental to enrich for potential responders to targeted therapies, this has been unsuccessful for chemotherapy, in part due to its often incompletely understood and diverse mechanisms of action.

Chemotherapy

The fluoropyrimidine antimetabolite 5-fluorouracil (5-FU) and leucovorin, a biomodulator that enhances 5-FU activity, are most often administered in combination with oxaliplatin, a platinum compound endowed with interstrand and intrastrand DNA cross-linking activity (FOLFOX; ref. 10), or irinotecan, a topoisomerase I inhibitor (FOLFIRI; ref. 11). Other therapeutic options include the fluoropyrimidine capecitabine plus oxaliplatin (CAPOX/XELOX) and capecitabine plus irinotecan (XELIRI). CAPOX/XELOX has shown analogous efficacy and safety compared with FOLFOX, and it is typically given as first- or second-line therapy in patients refractory to irinotecan-based chemotherapy (12). XELIRI is noninferior to FOLFIRI in terms of OS and is now recommended as an alternative second-line backbone treatment (13). In patients who have progressed after all standard therapies, a statistically significant (but modest) improvement in OS can be obtained with TAS-102, an agent that combines trifluridine (a nucleoside analogue) and tipiracil hydrochloride (an inhibitor of thymidine phosphorylase; ref. 14).

Potential determinants of response to chemotherapy have been brought to the fore based on the mechanism of action and metabolism of the various agents. However, the application of such predictors in clinical practice has been hampered by inconsistent results among different case series and poor diagnostic sensitivity and specificity. For some chemotherapeutics, in consonance with data from targeted therapies, drug target overexpression may be a positive determinant of sensitivity. For example, high expression of thymidylate synthase (TS), a direct target of 5-FU, has been associated with longer survival in patients with colorectal cancer treated with adjuvant 5-FU–based therapy in some studies (15, 16); however, other reports have not confirmed the positive predictive value of TS overexpression (refs. 17, 18; Table 1). Likewise, elevated levels of topoisomerase I appear to predict better response to irinotecan (ref. 19; Table 1). The activity of drug metabolic pathways is also thought to affect chemosensitivity. Dihydropyrimidine dehydrogenase (DPD) is a rate-limiting enzyme in fluoropyrimidine catabolism. High expression of DPD has been documented in tumors from patients with reduced sensitivity to capecitabine (20), with or without irinotecan (21), whereas inactivating polymorphisms of the DYPD gene (encoding DPD) have been associated with acute toxicity over the course of fluoropyrimidine-based therapy (refs. 22–24; Table 1). In the same vein, deleterious polymorphisms of the UGT1A1 gene (encoding glucuronosyltransferase, a key enzyme of irinotecan metabolism) are more frequent in patients who experience severe toxicity during treatment with irinotecan-based regimens (refs. 25, 26; Table 1).

Table 1.

Validated and proposed biomarkers of response to existing therapies in colorectal cancer

Validated and proposed biomarkers of response to existing therapies in colorectal cancer
Validated and proposed biomarkers of response to existing therapies in colorectal cancer

Responsiveness to chemotherapy may also be related to defects in DNA repair mechanisms after chemotherapy-induced DNA damage, leading to abnormalities in DNA replication and/or chromosome segregation that culminate in cancer cell death. Excision repair cross-complementation group 1 (ERCC1) is a key effector of DNA repair mechanisms and influences the tumor DNA–targeting effect of oxaliplatin. Some studies have shown that low transcript expression of the ERCC1 gene correlates with longer survival of patients treated with FOLFOX (ref. 27; Table 1). Similar findings were reported for an ERCC1 polymorphism at codon 118, which is expected to result in decreased ERCC1 gene expression (ref. 28; Table 1). These correlations, however, have not been confirmed in other datasets, especially when ERCC1 protein amounts rather than transcript expression were analyzed (29, 30). Functional studies are needed to deepen mechanistic investigation of the relationship between DNA repair deficiency and chemosensitization. More generally, we advocate a revamping of biologically oriented research as a means to disentangle the intricacies behind chemotherapy efficacy (or lack of), including the evaluation of how genetic and epigenetic modifications in components of DNA repair pathways shape response to cytotoxic agents. A clearer understanding of the cellular and molecular underpinnings of chemotherapy activity in clinically relevant experimental models is a necessary step for the nomination of response biomarkers above and beyond descriptive variables in patients.

EGFR mAbs

The EGFR mAbs cetuximab and panitumumab are currently used in association with FOLXOX or FOLFIRI in the first- or second-line treatment of patients with KRAS or NRAS wild-type tumors (31). The restricted use of anti-EGFR antibodies to patients with RAS wild-type mCRC is the result of a population-level biomarker-development strategy motivated by the plausible rationale that constitutive activation of signaling pathways downstream from EGFR—such as those triggered by RAS mutations—should bypass EGFR inhibition and therefore obviate sensitivity to EGFR-targeted agents. Evidence of the correlation between RAS genetic alterations and lack of response to EGFR blockade was initially limited to tumors with exon 2 KRAS mutations (32) and was later extended to KRAS exons 3 and 4 and NRAS exons 2, 3, and 4 (ref. 33; Table 1). The predictive value of the association was so strong that retrospective studies in patients were deemed sufficiently powered to guide the development of companion diagnostics for the routine assessment of “RAS extended” mutations in patients with mCRC, even in the absence of prospective validation.

Although patients with “RAS extended” mutations are currently excluded from therapy with cetuximab or panitumumab, there is still some debate as to whether KRASG13D mutations—different from all other RAS mutations—predict some benefit from EGFR antibody treatment. Based on retrospective pooled analyses of multiple trials, the addition of cetuximab to first-line or salvage chemotherapy was shown to improve the outcome of patients with KRASG13D-mutant tumors (with relative treatment effects similar to those observed in subjects with KRAS wild-type tumors, but with lower absolute values; refs. 34, 35). This association was not reproduced in another pooled retrospective evaluation of patients treated with panitumumab and chemotherapy (36). Whether this discrepancy is due to the different characteristics of the two antibodies (cetuximab is a human–mouse chimeric antibody that contains the human IgG1 constant region, whereas panitumumab is a fully humanized IgG2), or to differences in the clinical characteristics of the patient subgroups analyzed, remains to be determined. It is worth noting that the longer survival enjoyed by subjects with KRASG13D-mutant tumors treated with cetuximab and salvage chemotherapy was not observed in the cetuximab monotherapy arm (34), a finding confirmed in an independent study in patients who had received single-agent cetuximab (37); hence, the confounding effect of the chemotherapy backbone cannot be excluded. The potentially stronger reliance of cancer cells with KRASG13D mutations on the EGFR pathway has received some experimental support; specifically, KRASG13D-mutant cell lines have been demonstrated to be sensitive to the RAS-inhibitory activity of the GTPase-activating protein neurofibromin (NF1), which is possibly unleashed by EGFR blockade; this dependence of KRASG13D-mutant cells on the EGFR–NF1 axis could occur either because KRASG13D proteins are particularly responsive to NF1-stimulated GTP hydrolysis (38) or because they are incompetent to curb NF1-dependent inactivation of the wild-type KRAS allele product, which thus remains modulatable by upstream EGFR signaling (39). However, most cell lines with KRASG13D mutations also harbor NF1 loss-of-function alterations (38), so the proposed regulatory circuit is likely to be valid only in restricted model systems that are hardly representative of clinical reality.

Although the use of RAS mutation panels for the exclusion of patients with mCRC who will not benefit from anti-EGFR therapy is now commonplace, the quest for positive response predictors has lagged behind. EGFR is hardly ever mutated or amplified in colorectal cancer, indicating that tumor dependency on the EGFR pathway does not have an evident genetic basis. Retrospective correlative studies have documented higher tumor expression of the EGFR ligands amphiregulin and epiregulin in patients who respond to EGFR antibodies (refs. 40, 41; Table 1). Hence, mCRC tumors that are sensitive to EGFR neutralization appear to rely on EGFR signals owing to ligand-mediated autocrine or paracrine receptor activation. This knowledge has not translated into clinical-grade methods for selection of potential responders due to the difficulty of dichotomizing continuous variables, such as transcript or protein expression, into a digital cutoff for univocal allocation of patients to treatment.

Antiangiogenic Therapy

The first-line treatment for patients with mCRCs harboring KRAS or NRAS mutations comprises either FOLFIRI or FOLFOX plus the anti-VEGF antibody bevacizumab (42, 43). Whether the addition of VEGF-targeting agents to chemotherapy provides comparable or superior benefit to anti-EGFR antibodies in the context of KRAS/NRAS wild-type tumors is still a matter of debate (44). Beyond bevacizumab, two other antiangiogenic drugs have been proved to positively affect progression-free survival (PFS) and response rates when combined with chemotherapeutic agents in late lines of treatment: aflibercept (an anti-VEGFA, VEGFB, and placental growth factor; ref. 45) and ramucirumab (an anti-VEGFR2; ref. 46). Finally, heavily treated chemorefractory patients can experience slightly longer OS when treated with the multikinase inhibitor regorafenib, which also targets proangiogenic receptors (47).

At present, there are no validated predictors of response to antiangiogenic agents (48). Clinical reports have shown a correlation between more marked responsiveness to bevacizumab and low baseline levels of VEGFA splice isoforms (49), VEGFD (50), HGF (51), IL8 (52), or the VEGFA coreceptor neuropilin-1 (53), either in plasma or in tumors (Table 1). Germline polymorphisms of VEGFA (54, 55) and FLT1 (encoding VEGFR1; ref. 56), and inflammation- and endoplasmic reticulum–associated genes (57) also show a significant interaction with bevacizumab effectiveness (Table 1). Other potential biomarkers predicting bevacizumab therapeutic efficacy include loss of chromosome 18q11.2–q12.1 (58) and, more generally, a high degree of chromosomal instability (CIN; ref. 59; Table 1). In the case of regorafenib, initial studies suggest that low expression of vascular cell adhesion protein 1 (VCAM1) may be associated with better response (ref. 60; Table 1). Polymorphisms of genes related to the C-C motif chemokine ligand 5/C-C motif chemokine receptor 5 pathway (CCL5/CCR5; which regulates VEGFA expression) also predict efficacy in patients with mCRC treated with regorafenib (ref. 61; Table 1).

The fact that the expression levels of angiogenic targets positively associate with sensitivity to antiangiogenic agents has a pharmacokinetic basis ascribable to the relative stoichiometry of drug–target interactions. The biological significance of other candidate biomarkers, such as copy-number alterations (CNA), is of difficult interpretation in the absence of functional modeling in adequate experimental systems. Mechanistic investigation of stromal-directed therapies is complicated by a dearth of preclinical resources, which typically consist of syngeneic transplants, genetically modified mice, or xenografts. All approaches have limitations: On the one hand, “mouse-only” models hardly recapitulate the interpatient heterogeneity and population diversity of human tumors—a severe drawback for biomarker discovery research; on the other hand, xenografts are by definition a chimeric source of angiogenic factors, which can be concomitantly released by human cells of the tumor and murine stromal and inflammatory cells of the host. Therefore, due to species specificity, the cross-talk between heterologous cellular compartments is biased, and therapeutic antibodies cannot have full capacity at the organismal level because they selectively target either murine or human antigens.

Other Targeted Therapies

All the above therapies are administered in the absence of positive molecular selection; the only criterion is patient exclusion based on the presence of negative response biomarkers, as epitomized by the established association between KRAS/NRAS mutations and lack of response to EGFR blockade. Recently, a number of low-frequency aberrations in kinase-encoding genes have been identified in mCRC that result in constitutive activation of the corresponding protein products. Alterations in the ERRB2 gene (mostly gene amplification) are detected in around 5% of KRAS, NRAS, or BRAF wild-type mCRCs and lead to overexpression and hyperactivation of the encoded HER2 tyrosine kinase receptor. Phase II studies in patients selected for having HER2-positive mCRC tumors have shown that dual inhibition of HER2 using the anti-HER2 antibody trastuzumab and the EGFR/HER2 small-molecule inhibitor lapatinib (HERACLES trial) or the combination of trastuzumab and the anti-HER2 antibody pertuzumab (MyPathway trial) have considerable clinical efficacy, with around 30% response rates in heavily pretreated patients (refs. 62, 63; Table 1). Notably, in both studies, responsive patients had tumors with higher ERBB2 gene copy number than resistant patients, consistent with the assumption that higher ERBB2 gene dosage translates into stronger kinase activation, hence into more profound tumor dependency on HER2 signaling.

Kinase fusions originating from chromosomal translocations and resulting in constitutive activation of neurotrophic receptor tyrosine kinase 1 (NTRK1), NTRK2, NTRK3, anaplastic lymphoma kinase (ALK), and RET account for approximately 1% to 2% of KRAS, NRAS, or BRAF wild-type mCRCs. These rearrangements are enriched in right-sided RAS wild-type tumors, and though typically portending a dismal prognosis (64), they predict therapeutic benefit of inhibitors such as entrectinib (targeting NTRK, ROS1, and ALK; refs. 65, 66) and ponatinib (targeting various tyrosine kinases including RET; refs. 67, 68; Table 1).

BRAF gene alterations (with a dominant prevalence of V600E-activating mutations) are found in 7% to 10% of mCRCs (69, 70) and are generally mutually exclusive with KRAS and NRAS mutations, indicating that a single oncogenic hit on the ERK/MAPK pathway is sufficient to sustain tumorigenicity. Selective BRAF targeting with specific inhibitors has proved ineffective in patients with BRAF-mutant mCRC due to feedback reactivation of EGFR signaling, which substitutes for BRAF blockade in stimulating the MAPK pathway (71, 72). This observation has prompted the design of clinical trials aimed at evaluating the efficacy of combined BRAF and EGFR inhibition in patients with BRAF-mutant mCRC. In a recent phase III study testing cetuximab and the BRAF inhibitor encorafenib versus cetuximab and irinotecan (or FOLFIRI), combined EGFR and BRAF blockade significantly improved response rates and OS compared with standard therapy. This superior activity was further enhanced by concomitant MEK inhibition (ref. 73; Table 1).

Despite their high prevalence (approximately 50% of all colorectal cancers), KRAS- and NRAS-mutant tumors are still treated with conventional chemotherapy and antiangiogenic agents. Hopes are now placed on the use of KRASG12C covalent inhibitors, which are currently being tested in patients with KRASG12C solid tumors. Initial results seem to indicate that response rates to these drugs are relatively high in patients with non–small cell lung cancer but limited in patients with mCRC, likely due to retained sensitivity of colorectal cancer tumors to upstream EGFR signaling (ref. 74; Table 1). These observations echo findings in BRAF-mutant tumors (71, 72) and strengthen the notion that EGFR signaling needs to be concomitantly neutralized to achieve better responses to drugs targeting the RAS–MAPK pathway in mCRC.

Immunotherapy

About 15% of colorectal cancers display defective functionality of mismatch repair (MMR) proteins, which participate in the correction of base-pair mismatches occurring during DNA replication (especially at the level of repetitive DNA sequences called microsatellites). Deficient-MMR (dMMR) tumors with mutations in 30% or more microsatellites (defined as dMMR/MSI-H, i.e., with high microsatellite instability) tend to accumulate nonsynonymous mutations; this increased mutational burden can translate into a higher neoantigen load, which makes some dMMR/MSI-H tumors immunogenic and sensitive to immune checkpoint blockade (75). Accordingly, single-agent therapy with the anti–PD-1 antibodies pembrolizumab or nivolumab and combination therapy with nivolumab and the anti-CTLA4 antibody ipilimumab have been approved for treatment of patients with chemorefractory dMMR/MSI-H mCRC (refs. 76–78; Table 1). Preclinical evidence also suggests that anti–PD-1 immunotherapy may complement the activity of KRASG12C covalent inhibitors. In a colorectal cancer syngeneic mouse xenograft model dependent on the KRASG12C allele, KRASG12C blockade resulted in increased infiltration of CD8+ cytotoxic T cells, macrophages, and dendritic cells (79). This proinflammatory phenotype sensitized tumors to immunotherapy: When combined with an anti–PD-1 antibody, the KRASG12C inhibitor induced complete tumor regressions that persisted after treatment discontinuation. Interestingly, mice that were “cured” by the combined treatment against KRASG12C and PD-1 rejected tumor rechallenge, indicating that the combination therapy favored the establishment of tumor-specific T-cell responses (79).

The assessment of MSI status is now routinely performed for selecting patients likely to respond to immunotherapy, but only a subgroup of individuals with MSI mCRC receive clinical benefit from this treatment. The protein products of somatic DNA variants are not always efficiently presented by MHC molecules, which means that tumor mutational burden only partially contributes to neoantigen load. Likewise, although an association between high neoantigen load and pronounced immune cell infiltration has been repeatedly documented, the presence of an active immune microenvironment does not have predictive value for immunotherapy sensitivity (80). Human leukocyte antigen (HLA) binding prediction tools and artificial intelligence algorithms for multiparametric imaging of immune cell representation and topography in tumors are expected to yield more reliable molecular biomarkers for effective patient stratification in the immuno-oncology space.

Biomarkers that predict patient response to treatment are usually identified using population-based association studies, in which clinical outcome is correlated with a statistically significant enrichment for a specific molecular trait (typically, a genetic alteration) in subjects who do or do not respond to a given therapy. Albeit useful for clinical decision making, this approach fails to inform whether therapeutically relevant response predictors causally influence drug sensitivity and does not provide insight into the mechanistic underpinnings of the observed correlations. In a complementary perspective, studies using cancer cell lines enable extracting functional annotations and modeling cause–effect relationships; however, cell lines are by definition limited in number; thus, they do not recapitulate the spectrum of genetic heterogeneity spanned by patient tumors. Recently, patient-derived platforms that reflect the diversity of cancers, while retaining experimental manipulability and clinical fidelity, have been developed with the aim to characterize response biomarkers, investigate tumor adaptation under drug pressure, and understand the evolutionary principles of tumor progression. Colorectal cancer has been—and still is—a testing arena for such efforts.

Patient-Derived Xenografts for Validation of Targeted Therapy Biomarkers

Surgically derived tumor samples that are implanted in mice [known as patient-derived xenografts (PDX)] retain the inherent features of different tumors from different patients (6, 81). Vast PDX collections are therefore expected to capture interpatient tumor heterogeneity at the population level in a clinically relevant in vivo setting (82). Colorectal cancer is a paradigmatic example of the importance of PDX-based research for large-scale genotype–response associations, predictive biomarker identification, and therapeutic studies (refs. 83, 84; Fig. 1). In 2011, a systematic survey of KRAS and NRAS mutations in more than 100 PDXs from mCRC tumors, coupled with annotation of sensitivity to cetuximab, produced a dataset with both confirmatory and discovery aspects (85). On the one hand, the association between KRAS mutations in exon 2 and de novo resistance to EGFR blockade—which had emerged from clinical studies some years earlier (32, 86)—was “reverse validated” in PDXs and found to be coherent with patient data (ref. 85; Table 1). On the other hand, results in PDXs were among the first to illustrate that KRAS mutations in exons 3 and 4 and NRAS mutations predict lack of response to EGFR antibodies (85, 87). This finding would receive ultimate clinical recognition only 2 years later, when a retrospective–prospective analysis concluded that patients with tumors harboring “RAS extended” mutations treated with anti-EGFR antibodies had inferior PFS and OS compared with patients with KRAS/NRAS wild-type tumors (33).

Figure 1.

Application of colorectal cancer PDXs in translational research. PDX trials are conducted in parallel with or after clinical trials to “reverse validate” response biomarkers and genotype–response associations identified in patients (left). Cells dissociated from PDXs can be genetically manipulated and used in lineage-tracing experiments to assess the temporal and spatial dynamics of functionally heterogeneous clones under drug pressure (middle). Genomic analysis of large-scale PDX collections enables the discovery of molecularly defined colorectal cancer subpopulations, which can be tested for the presence of potential therapeutic targets through pharmacologic experiments in vivo (right). CAF, cancer-associated fibroblast.

Figure 1.

Application of colorectal cancer PDXs in translational research. PDX trials are conducted in parallel with or after clinical trials to “reverse validate” response biomarkers and genotype–response associations identified in patients (left). Cells dissociated from PDXs can be genetically manipulated and used in lineage-tracing experiments to assess the temporal and spatial dynamics of functionally heterogeneous clones under drug pressure (middle). Genomic analysis of large-scale PDX collections enables the discovery of molecularly defined colorectal cancer subpopulations, which can be tested for the presence of potential therapeutic targets through pharmacologic experiments in vivo (right). CAF, cancer-associated fibroblast.

Close modal

Although PDXs appear to have adequate predictive power for cancer cell–directed treatments, they lose value when dealing with therapies against stromal components—such as cancer-associated fibroblasts (CAF), endothelial cells, and inflammatory cells—and cells of the adaptive immune system. Indeed, the host must be immunocompromised to tolerate the graft, and human stromal cells are substituted with murine counterparts over serial passaging (6). But this drawback bears some advantages: The chimeric nature of PDXs has been leveraged to decompose—from bulk tumors—cancer cell–specific and stromal signals using analytic methods that distinguish human versus mouse transcripts. This exercise has increased the granularity and informative merit of gene expression classifications. For example, the clinical aggressiveness of a poor-prognosis transcriptional subtype named CMS4 had been initially ascribed to the ability of cancer cells to undergo epithelial–mesenchymal transition (EMT), a phenotypic switch that instigates cell motility and invasion (8, 88). With the possibility to discriminate between human and mouse transcripts, it became clear that—together with displaying some cancer cell–autonomous EMT traits—the vast majority of mesenchymal CMS4 tumors are in fact characterized by a heavy content of stromal cells, which likely foster the malignant characteristics of this subtype by conveying mitogenic, proinvasive, and antiapoptotic cues (89). In the same vein, Colorectal Cancer Intrinsic Subtypes (CRIS), a new colorectal cancer classification based only on PDX human transcripts, identified subtypes endowed with prognostic and predictive significance and showing limited overlap with transcriptional classes obtained from whole bulk colorectal cancers (90). Moreover, by focusing on cancer cell–intrinsic gene expression features that are not influenced by stromal abundance in isolated, randomly taken tumor samples, CRIS demonstrated higher accuracy in clustering colorectal cancers by patient of origin rather than tumor region of origin (91).

PDXs to Study the Clonal Dynamics of Colorectal Tumors under Chemotherapy Pressure

Tumors are composed of heterogeneous cell subsets that display different proliferation kinetics, susceptibility to apoptosis, and sensitivity to drug insults (92). Some works have used PDX models to investigate the clonal propagation dynamics of colorectal cancer subpopulations, both during spontaneous tumor growth and under drug pressure (Fig. 1). DNA CNA profiling and deep sequencing of mutational hotspots were combined with lentiviral lineage tracking to follow the progeny of single colorectal cancer cells over serial xenografts and to interrogate the relative contribution of genetic and nongenetic mechanisms to the functional heterogeneity of the individual cancer cells (93). Although genetically identical clones remained stable upon serial transplantation, lentivirally marked lineages were variable within each clone, with pronounced differences in proliferation rates, ability to persist, and susceptibility to exhaust through passages (93). Likewise, treatment of xenografts with oxaliplatin did not result in a detectable bottleneck or selection for novel genetic clones; rather, chemotherapy shaped a new dominance of previously dormant lineages and culled actively proliferating progeny (93). Together, these results indicate that cancer cell subpopulations can be genetically homogeneous (and stable) but functionally heterogeneous (and plastic) in colorectal cancer.

The finding that colorectal cancer cells oscillate between periods of dormancy and activity appears to have a positional determination. Using a tamoxifen-inducible labeling system to stochastically mark cancer cells in mouse xenografts of patient-derived spheroids, coupled with computational modeling, Lenos and colleagues documented that colorectal cancer grows through surface expansion (94). This peripheral accretion is driven by the local availability of mitogenic gradients secreted by CAFs, which are sensed only by cancer cells located in the outermost zone of the tumor (ref. 94; Fig. 1). Chemotherapy with 5-FU and oxaliplatin reduced tumor growth rates but did not affect the residual dynamics of surface growth, indicating that microenvironmental influences dictating spatially confined cell proliferation are not interfered with by cytotoxic treatment (94). Other studies with multicolor lineage tracing approaches in xenografts of colorectal cancer primary cultures and cell lines confirmed that tumor outgrowth is geometrically orchestrated by large proliferating clones confined at the leading tumor edge, whereas small quiescent clones reside in the center (95, 96).

PDXs and Genetically Modified Animal Models for Target Discovery

Besides providing preclinical hints for response biomarker validation, PDXs have also been deployed for testing therapeutic options in newly identified, molecularly circumscribed mCRC subsets (Fig. 1). Amplification of the ERBB2 oncogene was detected in some KRAS/NRAS wild-type, cetuximab-resistant PDX models, and was found to predict response to HER2-targeted therapies in PDX-based preclinical trials (refs. 85, 97; Table 1). Other clinically actionable alterations were shown to be enriched in KRAS/NRAS wild-type, cetuximab-refractory PDXs and patients, including activating mutations of ERBB2 and MAP2K1 (encoding the RAS downstream effector MEK1), amplification of the tyrosine kinase receptors MET and FGFR1, and outlier overexpression of the survival factor IGF2 (98–101). In general, the sole inhibition of the hyperactive oncoproteins proved to be ineffective in PDXs, but treated tumors were invariably sensitized to concomitant EGFR blockade. Subsequent clinical studies confirmed that patients with HER2-positive mCRC tend to respond poorly to EGFR antibodies and can benefit from dual treatments against HER2 and EGFR (62, 63). Similarly, MEK1 mutations were found to predict resistance to EGFR inhibition and response to a combination of trametinib (a MEK inhibitor) and panitumumab in patients (102).

PDXs have also been shown to recapitulate clinical reality in terms of depth of response. Similar to metastases in patients, mCRC PDXs that respond to EGFR antibodies can experience massive shrinkage but are hardly ever eradicated. The residual cancer cells that withstand up-front drug treatments act as a reservoir for the stochastic acquisition of resistance-conferring mutations, with the ensuing expansion of subclones responsible for tumor relapse (103). Recent evidence indicates that residual mCRC PDXs (and residual tumors in patients) at maximal response to prolonged anti-EGFR therapy relax their dependency on EGFR signals by reducing the expression of genes encoding EGFR-activating ligands and increasing alternate HER2/HER3 pathway activity, while becoming similar to slowly cycling secretory precursors of the normal intestine (104). The finding that cetuximab-tolerant residual tumors exhibit decreased abundance of EGFR cognate ligands is consistent with the clinical observation that patients with mCRC tumors expressing low levels of amphiregulin and epiregulin tend to respond less to EGFR antibodies (refs. 40, 41; Table 1). Pseudodifferentiation into tissue-specific lineages has been documented as a mechanism of therapy resistance in other tumors; for example, the manifestation of neuroendocrine traits is a hallmark of emerging resistance to EGFR inhibitors and antiandrogen treatment in lung and prostate cancers, respectively (105, 106). In the context of colorectal cancer, cetuximab-induced phenotypic reprogramming toward a secretory fate with high HER2/HER3 signaling makes cancer cells vulnerable to concomitant targeting of EGFR, HER2, and HER3, as shown by reduction of residual disease burden and prolonged time to relapse after treatment discontinuation in PDX trials with a pan-HER antibody (104).

As noted above, PDXs are inadequate tools for predicting response to therapies against stromal and immune cells. Genetically modified mouse models (GEMM) develop autochthonous colorectal cancer tumors in an immune-competent background, but the artificial introduction of founder oncogenic mutations may result in evolutionary trajectories different from those occurring in spontaneous tumors (107). This limitation has been addressed by engineering the ordered expression of salient mutant oncoproteins along the linear progression sequence that typifies human colorectal cancer, with the aim to more faithfully recapitulate the natural history of human tumors. In seminal experiments, individual inactivation of the Apc gene (which normally represses intestinal stem cell proliferation by blocking mitogenic signals of the WNT pathway) caused the formation of adenomas and in situ carcinomas (108). In the context of Apc deficiency, the concomitant expression of oncogenic Kras or the concomitant loss of the tumor-suppressor gene Trp53 or the Smad2/Smad4 genes (the latter being downstream mediators of TGFβ signaling) accelerated intestinal tumorigenesis and resulted in the development of locally invasive (albeit not metastatic) carcinomas (109–112). Ultimately, the compound assortment of mutations in Apc, Kras, and Trp53 or Apc, Kras, and Tgfbr2 (encoding the type 2 TGFβ receptor) enabled implementation of the full metastatic phenotype (113, 114). Likewise, animals with targeted gene recombination of Apc, Kras, Tgfbr2, and Trp53 (AKTP) to intestinal stem cells developed invasive colorectal adenocarcinomas with hallmarks of human microsatellite-stable tumors, including low mutational burden and scant T-cell infiltration (115). Moreover, AKTP tumors had an abundant representation of CAFs engaged in massive deposition of extracellular matrix and profuse secretion of TGFβ. Importantly, increased TGFβ in the tumor microenvironment was found to be a major determinant of T-cell exclusion, and blockade of TGFβ signaling rendered tumors more T cell–inflamed and susceptible to immunotherapy (ref. 115; Fig. 2). Another mouse model harboring Kras and Trp53 mutations along with hyperactive Notch signaling in intestinal cells developed metastatic tumors with serrated morphology, extensive stromal content, and gene expression profiles similar to those of poor-prognosis tumors in patients (116). In these mouse tumors, hyperactivation of the Notch pathway resulted in secretion of TGFβ by cancer cells, which prompted neutrophil accumulation in the tumor stroma and neutrophil-dependent metastatic dissemination. Accordingly, targeting neutrophil recruitment or TGFβ signaling reduced metastatic burden (ref. 116; Fig. 2). Collectively, these results underscore the value of transgenic mice as investigational models to explore the interplay between genetic alterations and the immune-competent tumor microenvironment and to integrate PDX-based research on cancer cell–intrinsic vulnerabilities.

Figure 2.

Application of genetically modified animal models of colorectal cancer in translational research. Genetically modified mice carrying targeted gene recombination of common mutations (Apc, Kras, Tgfbr2, and Trp53) in intestinal stem cells develop immune-cold colorectal tumors with high levels of stromal TGFβ; blockade of TGFβ signals prompts the recruitment of immune effector cells into the tumor microenvironment and sensitizes tumors to immunotherapy (left). Another mouse model develops metastatic colorectal cancer featuring a pronounced stromal reaction due to targeted expression of active Kras and Notch and loss of Trp53 in Villin-positive intestinal cells; Notch-dependent production of TGFβ by cancer cells promotes tumor infiltration by neutrophils and metastatic dissemination, which can be blunted by inhibition of neutrophil recruitment or TGFβ signaling (right).

Figure 2.

Application of genetically modified animal models of colorectal cancer in translational research. Genetically modified mice carrying targeted gene recombination of common mutations (Apc, Kras, Tgfbr2, and Trp53) in intestinal stem cells develop immune-cold colorectal tumors with high levels of stromal TGFβ; blockade of TGFβ signals prompts the recruitment of immune effector cells into the tumor microenvironment and sensitizes tumors to immunotherapy (left). Another mouse model develops metastatic colorectal cancer featuring a pronounced stromal reaction due to targeted expression of active Kras and Notch and loss of Trp53 in Villin-positive intestinal cells; Notch-dependent production of TGFβ by cancer cells promotes tumor infiltration by neutrophils and metastatic dissemination, which can be blunted by inhibition of neutrophil recruitment or TGFβ signaling (right).

Close modal

Patient-Derived Organoids for Mechanistic Investigation and Pharmacologic Studies

PDXs represent more authentic working models than conventional cell line xenografts to study how cancer cells evolve and react to therapies in a clinically relevant scenario that reflects organismal complexity. However, PDXs are not endowed with sufficient experimental tractability to distill causality from description, nor do they show sufficient scalability to enable high-throughput pharmacogenomic screens. Short-term culture of tumor sections allows for in vitro screening at a reasonably large scale (117), but it is constrained by the fact that the proliferative capacity of the cultures dissipates over time. To overcome these limitations, three-dimensional organotypic or “organoid” long-term culture methods have been developed that combine the architectural complexity of tissues with the experimental flexibility of “immortalized” cell culture systems (118). For the colon, normal organoids containing only epithelial cells can be derived by culturing primary nontransformed intestinal tissue in Matrigel—a gelatinous mixture made of laminin-rich extracellular matrix and growth factors—supplemented with additional growth factors that mimic the intestinal niche (refs. 119, 120; Fig. 3). Colorectal cancer organoids require less stringent combinations of niche factors than normal intestinal organoids (121, 122). Mouse and human organoids are commonly used not only for biological and pharmacologic studies in vitro but also as model tools of colorectal cancer spontaneous metastasization. GEMM-derived organoids, normal mouse organoids engineered to express oncogenic mutations, and patient-derived human tumor organoids can readily give rise to invasive carcinomas that infiltrate the muscularis propria and colonize the liver after orthotopic engraftment by colonoscopy-guided mucosal injection, enema, or surgical implantation into the submucosa of the caecal wall (123–127).

Figure 3.

Application of colorectal cancer (CRC) organoids in translation research. Patient-derived organoids from normal colon can be engineered to express drivers of colorectal tumorigenesis, alone and in combination; this approach allows us to explore the contribution of each driver to tumor onset and progression and helps us understand how and to what extent engineered organoid models recapitulate the biological characteristics of spontaneous tumors from patients (left). Mutational profiles, methylomics, and/or RNA-sequencing analysis of clonal organoids derived from individual cells of patients' tumors can be used to reconstruct phylogenetic trees and investigate colorectal cancer tumor evolution (middle). Organoids can be exploited in mid- to high-throughput drug screens, and results from pharmacologic analyses can be coupled with molecular profiles to extract associations between drug sensitivity and specific molecular traits (right).

Figure 3.

Application of colorectal cancer (CRC) organoids in translation research. Patient-derived organoids from normal colon can be engineered to express drivers of colorectal tumorigenesis, alone and in combination; this approach allows us to explore the contribution of each driver to tumor onset and progression and helps us understand how and to what extent engineered organoid models recapitulate the biological characteristics of spontaneous tumors from patients (left). Mutational profiles, methylomics, and/or RNA-sequencing analysis of clonal organoids derived from individual cells of patients' tumors can be used to reconstruct phylogenetic trees and investigate colorectal cancer tumor evolution (middle). Organoids can be exploited in mid- to high-throughput drug screens, and results from pharmacologic analyses can be coupled with molecular profiles to extract associations between drug sensitivity and specific molecular traits (right).

Close modal

Patient-derived normal and colorectal cancer organoids have been leveraged to advance cancer modeling and decompose mechanisms of colorectal cancer tumorigenesis. Using CRISPR/Cas9-based genome editing, Matano and colleagues sequentially introduced loss-of-function mutations of APC, SMAD4, and TP53 and gain-of-function mutations of KRAS and/or PIK3CA in normal human colon organoids, followed by growth selection under customized cell culture conditions (128). Organoids engineered to express all five mutations formed small, highly differentiated tumors with limited local infiltration after implantation under the kidney subcapsule in mice, and were unable to metastasize to the liver after injection into the spleen. Conversely, organoids from patients' advanced tumors that had accumulated spontaneous oncogenic mutations during their evolutionary history displayed robust renal subcapsular growth and produced prominent spleen-to-liver dissemination (ref. 128; Fig. 3). These results suggest that the ectopic introduction of canonical driver mutations in normal human intestinal cells results in incipient tumor formation but is not sufficient for a colorectal cancer tumor to exhibit an invasive and metastatic behavior. Additional lesions that drive full-blown colorectal cancer malignancy may be fueled by epigenetic modifications and CIN; indeed, the engineered organoids largely lacked karyotypic or DNA methylation aberrations, which were instead abundantly present in patient-derived colorectal cancer organoids (128). The acquisition of gene CNAs after genetic manipulation of normal intestinal organoids appears to be influenced by experimental variables; for example, different from Matano and colleagues, Drost and colleagues found that combined loss of APC and TP53 in normal human colon organoids was sufficient for the appearance of CIN and massive aneuploidy (129). In patient-derived colorectal cancer organoids, a combination of genetic lineage tracing and ablation systems revealed robust functional plasticity. LGR5+ cells were shown to act as cancer stem cells that constantly fueled tumor growth through self-renewal and at the same time were able to morph into differentiated postmitotic cells. Selective ablation of LGR5+ cells transiently regressed tumors; however, this shrinkage was followed by tumor regrowth due to the replenishment of the LGR5+ pool by differentiated cells that had reacquired stem-like features (130).

Clonal organoids derived from isolated cells can be considered as proxies for the single cells from which they originate (Fig. 3). Phylogenetic trees constructed through deep genomic analysis of colorectal cancer clonal organoids revealed that driver mutations commonly found in colorectal cancer (such as those in APC, KRAS, and TP53) were present in all organoids; that is, they were trunk mutations common to all cells of the original tumor. However, many “private” mutations could be detected in the distal branches of the phylogenetic trees, indicating that they had arisen later during tumor progression and had contributed to tumor genetic diversification (131). These results are in line with the “big bang” model of colorectal cancer tumorigenesis, according to which genetic variants that confer selective advantages occur early in a cancer's evolution and are followed by the neutral expansion of genetically different but equally fit subclones (132, 133). Stable alterations of DNA methylation and transcriptome states were also observed in clonal organoids, with phylogenetic topologies similar to the mutation-based trees (Fig. 3). Conversely, response to drugs commonly used in colorectal cancer was variable—especially with chemotherapeutic agents—and not linked to the geographic location of the organoid-initiating cells in the original tumor or the genetic distance between clones (131). Similar to those observed in PDX-based lineage tracking experiments (93), these results suggest that diversification in biological behavior has no evident correlation with the extent of mutational diversification.

The application of organoid technology in systematic high-throughput drug screens to validate clinically relevant response biomarkers and nominate new ones is rapidly expanding (Fig. 3). A seminal study with a library of 83 compounds tested in 19 organoids from primary colorectal cancer tumors confirmed the association between KRAS mutations and lack of response to EGFR blockade as well as general refractoriness of BRAF-mutant tumors to BRAF inhibitors, as observed in the clinic (121). This effort also identified loss-of-function mutations in RNF43, resulting in cell hypersensitivity to secreted WNT factors, as predictive biomarkers of colorectal cancer susceptibility to neutralization of autocrine/paracrine activation of the WNT pathway (121). Organoids from metastatic samples have been shown to recapitulate the clinical response of the donor patient to cetuximab, regorafenib, and TAS-102 (134). Interestingly, organoids derived from a patient with regorafenib-sensitive liver metastases proved to be resistant to the drug when cultured ex vivo as isolated cancer cells; however, liver orthotopic xenografts developed from the same organoids co-opted the host's blood vessels and displayed reduced vascularity after regorafenib administration, in keeping with the assumption that response to regorafenib is mainly driven by its antiangiogenic activity (134). A concordance between cancer cell viability and patient response was also found in organoids from metastatic tumors treated with irinotecan monotherapy or FOLFIRI, but not when FOLFOX was used (135). Possibly, stromal and immune components absent in organoid cultures tune sensitivity to oxaliplatin more than they do with other drugs, or reliable response to oxaliplatin requires tailored culture conditions that are less stringent for other chemotherapeutics. Finally, organoids derived from primary rectal cancers have been demonstrated to predict clinical and histopathologic responses to neoadjuvant chemoradiation, as observed in matched donor patients (136, 137).

The utilization of living biobanks of tumor samples holds considerable promise for in vivo and in vitro interrogation of clinically actionable pathways and for the study of tumor evolution. But the use of patient-derived models should be accompanied by careful appreciation of their real potential not only as platforms for biomarker validation and target discovery but also as reliable proxies of the biological and molecular fingerprints of matched tumors in donor patients. A critical knowledge of the accuracy of patient-derived models in retaining the characteristics of original tumors is crucial for assessing their ability to predict drug activity in the clinic.

Preservation of Genomic Architecture in Propagated Tumor-Derived Models

An ongoing debate revolves around the question of whether serially passaged PDXs and long-cultured organoids preserve the genomic makeup, in terms of CNAs, of their prederivation counterparts. Using gene expression microarray data to infer large-scale CNA profiles, Ben-David and colleagues reported extensive copy-number divergence between the preimplantation tumor of origin and the corresponding xenograft at the first in vivo passage, which was exacerbated along serial propagations (138). This raised concerns that mouse-specific selective pressures may “artificially” influence PDX tumor evolution, with implications for the ability of PDXs to faithfully model patient treatment response. However, expression-based CNA calling only enables assessment of aberrations at the gross scale of chromosomal arms. Recently, a joint effort of the National Cancer Institute PDXNet consortium and the EurOPDX consortium produced a DNA-based enumeration of copy-number profiles at high segmental resolution in a large collection of PDX models (139). This analysis did not confirm systematic copy-number deviation between patient tumors and PDXs; rather, it documented high CNA retention during PDX engraftment and passaging (both globally and at the level of cancer-related genes) for many tumor types including mCRC. Notably, CNA variations between preimplantation and xenografted tumors were comparable to differences in multiregion samples of tumors in patients, indicating that the impact of PDX-associated CNA drift is similar to the natural intratumoral evolution that occurs in patients.

Somatic mutations, typically assessed by whole-exome sequencing, are largely concordant between original tumors and matched PDXs, even though evolutionary neutral subclonal alterations may arise at low allele frequency during PDX propagation (140). In colorectal cancer, mutations in known oncogenic drivers are retained in PDXs when present in the corresponding patient tumors and do not appear de novo in mouse-passaged xenografts from either primary (84) or metastatic samples (85, 99), including matched samples of primary tumors and synchronous or metachronous metastases (83, 141). An overall preservation of CNA and mutational landscape, with the caveat that the number of samples analyzed so far is limited, has also been observed in colorectal cancer organoids as compared with the corresponding patient tumors (84, 121, 142). However, CIN colorectal cancer organoids tend to tolerate mitotic errors, which results in the accrual of chromosome missegregations over time (143). Similarly, an accumulation of synonymous and nonsynonymous mutations has been noted during prolonged culturing of MSI colorectal cancer organoids (122).

The Hurdles of Coclinical Trials

A number of exploratory studies have shown the potential of PDXs for mirroring therapeutic response in the patients who contributed tumor samples; for instance, the clinical outcome of individuals who had received various chemotherapeutic regimens for the treatment of liver or peritoneal metastases reflected the objective response (or lack of it) monitored months or years before in patient-matched PDXs that had been established at the time of primary tumor resection (144). Similarly, when PDX models were generated from pretreatment core biopsies of BRAF-mutant metastases and tested for their sensitivity to combined BRAF and MEK blockade, the objective response in mice was similar to the radiologic response in the biopsied lesions (145).

If patient-derived models are high-fidelity “avatars” of pre-derivation tumor samples, they could be used—in principle—for real-time assessment of drug sensitivity, which may be reverse-exploited to guide treatment decisions in donor patients. Coclinical trials have been proposed in which PDX mice are treated with panels of drugs—either agents with broad-brush anticancer activity or targeted compounds based on molecular predictors; then, when a positive signal for a specific therapy emerges, the information is transferred back to the donor patient for clinical evaluation (81, 146). Although intriguing, an approach of this kind requires that therapeutic findings be univocally deciphered and rigorously interpreted. For example, spurious positive signals may arise for treatments that delay tumor growth, resulting in tumors that are smaller than untreated controls at end point, but larger than they were at treatment initiation. This outcome may be indicative of biological sensitivity (i.e., the drug reduces cancer cell proliferation) but has little clinical relevance; indeed, in patients, a lesion that enlarges during treatment (even to a relatively small extent) denotes tumor progression, and the therapy is usually discontinued due to lack of efficacy.

Another issue with the execution of PDX-based coclinical trials is the need to cope with quick turnarounds. Results in mice must be promptly returned to donor patients to inform treatment decisions, but research with PDXs notoriously implies long-term and time-consuming experiments. “Cutting corners” in the name of rapidity, for example by reducing the number of animals tested in each treatment cohort, would lead to insufficiently powered studies and scientifically unreliable conclusions. Compared with PDXs, organoids are expected to speed up the bench-to-bedside pipeline due to their higher manageability. However, we are still missing metrics that adequately capture how and to what extent organoids deliver a clear prediction of the outcome in patients. There is no consensus on the adoption of common readouts of drug sensitivity (reduction of cell proliferation vs. induction of apoptosis) and shared methodologies for data acquisition (digital imaging vs. cell counts). Moreover, a direct comparison of the concordance between patient-matched PDXs and organoids in categorizing response or resistance to therapy has not been attempted so far on a systematic scale.

PDX studies could also provide potentially useful real-time information about drug toxicity, but gathering generalizable data on this aspect will likely prove daunting. A meta-analysis of adverse events in mice treated with various therapies has revealed large deviations among different studies, with a variable extent of animal weight loss or death toll that was apparently independent of mouse strain and dosage and rather attributable to facility- and operator-related factors (147). This inconsistency is compounded by idiosyncratic liabilities of defined mouse strains; for instance, SCID mice harbor a loss-of-function mutation in the catalytic subunit of DNA-dependent protein kinase, an enzyme required for efficient DNA double-strand break repair (148). Consequently, mouse strains that carry this mutation show increased total-body sensitivity to chemical or physical agents that damage DNA, such as irradiation, chemotherapy, and inhibitors of the DNA repair machinery. As always when dealing with resource platforms, standardized guidelines built on cumulative experience will be a prerequisite for direct transfer of preclinical results to patients.

Integration of the Tumor Immune Microenvironment: Humanized Mice and Cocultures

The necessity of recurring to immunocompromised mice to prevent xenograft rejection hampers the use of conventional PDX models to assess the efficacy of immunotherapies. Humanized mice are immunodeficient animals in which the human immune system is partially reconstituted by introducing CD34+ hematopoietic stem cells (HSC), peripheral blood mononuclear cells (PBMC), or tumor-infiltrating lymphocytes (TIL; refs. 6, 149; Fig. 4). Attempts to generate humanized colorectal cancer models have been scant. Cell line xenografts in mice engrafted with allogeneic or autologous human PBMCs showed delayed growth kinetics and increased infiltration of cytotoxic T cells after treatment with a combination of nivolumab and urelumab, a CD137 agonist mAb that enhances T cell and natural killer cell antitumor activity (150). Similar results were observed in a dMMR/MSI-H PDX model, but not in a microsatellite-stable model, after humanization with cord blood–derived CD34+ cells and treatment with nivolumab (151).

Figure 4.

Incorporating the immune system into patient-derived models. The immune system of immunocompromised mice can be partially reconstituted with different approaches of variable efficacy, from infusions of PBMCs or TILs to transplantation of HSCs derived from the bone marrow or umbilical cord blood; once humanized, mice can be xenografted with patient-derived tumors and treated with immunotherapy to assess tumor growth kinetics and intratumor representation of immune cells before and after treatment (left). Cocultures of immune cells and cancer cells can be performed by comingling tumor organoids and autologous PBMCs or by implementing air–liquid interface (ALI) methods that allow the preservation of the tumor epithelium and the associated immune stroma in cohesive units; both approaches are permissive for expansion of tumor-specific cytotoxic T cells (right). CRC, colorectal cancer; NK, natural killer.

Figure 4.

Incorporating the immune system into patient-derived models. The immune system of immunocompromised mice can be partially reconstituted with different approaches of variable efficacy, from infusions of PBMCs or TILs to transplantation of HSCs derived from the bone marrow or umbilical cord blood; once humanized, mice can be xenografted with patient-derived tumors and treated with immunotherapy to assess tumor growth kinetics and intratumor representation of immune cells before and after treatment (left). Cocultures of immune cells and cancer cells can be performed by comingling tumor organoids and autologous PBMCs or by implementing air–liquid interface (ALI) methods that allow the preservation of the tumor epithelium and the associated immune stroma in cohesive units; both approaches are permissive for expansion of tumor-specific cytotoxic T cells (right). CRC, colorectal cancer; NK, natural killer.

Close modal

Although humanized mice appear to recapitulate some of the effects of immunotherapy observed in patients, the procedure of mouse humanization is afflicted with several drawbacks. PBMC and TIL infusion typically causes severe graft-versus-host disease starting 2 to 5 weeks after injection (152, 153), which restricts the investigative window to temporal limits that are hardly compatible with experimental needs. Transplantation of HSCs results in a more complete hematopoietic reconstitution, but their maturation as well as the effector functions of their differentiated progeny are compromised by the lack of cytokines, phagocytes, and HLA molecules of human origin in the mouse host. The application of genome-editing technologies for mouse genetic engineering is expected to increase the extent of humanized cells and molecules in future murine models.

Another emerging asset to reconstruct the functional interactions between cancer cells and the immune microenvironment relies on hybrid organ-on-a-chip platforms, which allow the buildup of more complex multicellular systems (154). Reductionist methodologies involve the initial establishment of separate cultures of epithelial organoids and immune cells, followed by artificial reconstitution in comingling experiments. This approach has been used to set up cocultures of cancer cells from primary or metastatic colorectal cancer with high mutational burden and autologous PBMCs, wherein cancer cell organoids triggered antigen-specific stimulation of tumor-reactive cytotoxic T cells in the PBMC fraction (ref. 155; Fig. 4). More sophisticated air–liquid interface (ALI) methods have also been deployed that enable the en bloc preservation of the tumor epithelium and its endogenous immune stroma, including fibroblasts, tumor-associated macrophages, T and B lymphocytes, and natural killer cells (156). ALI cohesive units propagated from colorectal cancer biopsies retained the T-cell receptor heterogeneity of the T cells present in original tumors and modeled the effects of nivolumab by recapitulating cytotoxic T-cell expansion and antibody-dependent tumor cytotoxicity (ref. 156; Fig. 4). Further complexity could be achieved by integrating on-chip tumor immune microenvironments with biomimetic vascular-like structures for reconstitution of physiologic functions of the microvascular tissue. This methodology has been used to develop a three-dimensional chip-based model comprising a human colorectal cancer core and a surrounding vascularized network (157) and, in principle, might be upscaled to include microfluidic cocultures of immune cells. Assessing the functional consequences of immune checkpoint blockade using advanced organoid technology is poised to complement existing descriptive biomarkers, such as neoantigen load, in the identification of patient-specific determinants of response to immunotherapy.

The clinical and experimental observations discussed above illustrate the power of population-level studies—both in patients and in the preclinical setting—to credential candidate predictive biomarkers and identify novel determinants of therapeutic response as well as novel targets. Recent evidence also highlights the value of PDXs and organoids as tools to investigate subclonal dynamics during tumor evolution and functional heterogeneity under drug pressure. The credibility of patient-derived models in preserving the molecular architecture of the corresponding prederivation tumors is now supported by large-scale analytic efforts and the use of accurate genomic approaches. These merits notwithstanding, several issues remain, which are mostly related to the inability of PDXs and organoids to recapitulate heterotypic interactions between cancer cells, stromal cells, and immune cells. Mouse humanization procedures and coculture assays are expected to aid the development of more holistic models that incorporate immune components. However, the impact of bone marrow reconstitution (let alone that of PBMC or TIL infusion) on the quality, quantity, and topographic localization of immune infiltrates in transplanted tumors is difficult to assess, as is the influence of the host on the differentiation trajectories and functionality of transplanted human HSCs. Likewise, cocultures of cancer cell organoids with endogenous, syngeneic immune cells fail to mimic the subtleties of the tumor microenvironment in terms of complexity, representation, and reciprocal distribution of immune components. Another dimension of complexity is the difficulty—if not the impossibility—of replacing stromal elements such as endothelial cells and fibroblasts with their human counterparts; hence, the limitation remains that mouse-derived cytokines and growth factors in some cases do not cross-react with receptors that are expressed by human cancer cells.

A careful appraisal of the (vast) extent of information that can be reliably garnered by the use of patient-derived models, but also a clear understanding of their shortcomings, will be key to deliver robust, predictive, and translationally relevant knowledge. This critical attitude will help triage and move to the clinic only those findings that emerge from conclusive and generalizable preclinical research and are motivated by responsible and limitation-aware methodologic considerations.

L. Trusolino reports grants from Symphogen, Servier, Pfizer, Menarini, and Merus, and personal fees from Eli Lilly, AstraZeneca, and Merck outside the submitted work. No disclosures were reported by the other author.

This study was supported by Associazione Italiana per la Ricerca sul Cancro (AIRC) Investigator Grant 22802, AIRC 5 × 1000 grant 21091, AIRC/Cancer Research UK/FC AECC Accelerator Award 22795, H2020 grant agreement no. 754923 COLOSSUS, and Fondazione Piemontese per la Ricerca sul Cancro-ONLUS 5 × 1000 Ministero della Salute 2014, 2015, and 2016. L. Trusolino is a member of the EurOPDX Consortium.

1.
Siegel
RL
,
Miller
KD
,
Jemal
A
. 
Cancer statistics, 2020
.
CA Cancer J Clin
2020
;
70
:
7
30
.
2.
Ferlay
J
,
Colombet
M
,
Soerjomataram
I
,
Dyba
T
,
Randi
G
,
Bettio
M
, et al
Cancer incidence and mortality patterns in Europe: estimates for 40 countries and 25 major cancers in 2018
.
Eur J Cancer
2018
;
103
:
356
87
.
3.
Advani
S
,
Kopetz
S
. 
Ongoing and future directions in the management of metastatic colorectal cancer: update on clinical trials
.
J Surg Oncol
2019
;
119
:
642
52
.
4.
Kopetz
S
. 
New therapies and insights into the changing landscape of colorectal cancer
.
Nat Rev Gastroenterol Hepatol
2019
;
16
:
79
80
.
5.
Network CGA
. 
Comprehensive molecular characterization of human colon and rectal cancer
.
Nature
2012
;
487
:
330
7
.
6.
Byrne
AT
,
Alférez
DG
,
Amant
F
,
Annibali
D
,
Arribas
J
,
Biankin
AV
, et al
Interrogating open issues in cancer precision medicine with patient-derived xenografts
.
Nat Rev Cancer
2017
;
17
:
254
68
.
7.
Sasaki
N
,
Clevers
H
. 
Studying cellular heterogeneity and drug sensitivity in colorectal cancer using organoid technology
.
Curr Opin Genet Dev
2018
;
52
:
117
22
.
8.
Dienstmann
R
,
Vermeulen
L
,
Guinney
J
,
Kopetz
S
,
Tejpar
S
,
Tabernero
J
. 
Consensus molecular subtypes and the evolution of precision medicine in colorectal cancer
.
Nat Rev Cancer
2017
;
17
:
79
92
.
9.
Punt
CJ
,
Koopman
M
,
Vermeulen
L
. 
From tumour heterogeneity to advances in precision treatment of colorectal cancer
.
Nat Rev Clin Oncol
2017
;
14
:
235
46
.
10.
de Gramont
A
,
Figer
A
,
Seymour
M
,
Homerin
M
,
Hmissi
A
,
Cassidy
J
, et al
Leucovorin and fluorouracil with or without oxaliplatin as first-line treatment in advanced colorectal cancer
.
J Clin Oncol
2000
;
18
:
2938
47
.
11.
Douillard
JY
,
Cunningham
D
,
Roth
AD
,
Navarro
M
,
James
RD
,
Karasek
P
, et al
Irinotecan combined with fluorouracil compared with fluorouracil alone as first-line treatment for metastatic colorectal cancer: a multicentre randomised trial
.
Lancet
2000
;
355
:
1041
7
.
12.
Arkenau
HT
,
Arnold
D
,
Cassidy
J
,
Diaz-Rubio
E
,
Douillard
JY
,
Hochster
H
, et al
Efficacy of oxaliplatin plus capecitabine or infusional fluorouracil/leucovorin in patients with metastatic colorectal cancer: a pooled analysis of randomized trials
.
J Clin Oncol
2008
;
26
:
5910
7
.
13.
Xu
RH
,
Muro
K
,
Morita
S
,
Iwasa
S
,
Han
SW
,
Wang
W
, et al
Modified XELIRI (capecitabine plus irinotecan) versus FOLFIRI (leucovorin, fluorouracil, and irinotecan), both either with or without bevacizumab, as second-line therapy for metastatic colorectal cancer (AXEPT): a multicentre, open-label, randomised, non-inferiority, phase 3 trial
.
Lancet Oncol
2018
;
19
:
660
71
.
14.
Mayer
RJ
,
Van Cutsem
E
,
Falcone
A
,
Yoshino
T
,
Garcia-Carbonero
R
,
Mizunuma
N
, et al
Randomized trial of TAS-102 for refractory metastatic colorectal cancer
.
N Engl J Med
2015
;
372
:
1909
19
.
15.
Edler
D
,
Glimelius
B
,
Hallström
M
,
Jakobsen
A
,
Johnston
PG
,
Magnusson
I
, et al
Thymidylate synthase expression in colorectal cancer: a prognostic and predictive marker of benefit from adjuvant fluorouracil-based chemotherapy
.
J Clin Oncol
2002
;
20
:
1721
8
.
16.
Kornmann
M
,
Schwabe
W
,
Sander
S
,
Kron
M
,
Sträter
J
,
Polat
S
, et al
Thymidylate synthase and dihydropyrimidine dehydrogenase mRNA expression levels: predictors for survival in colorectal cancer patients receiving adjuvant 5-fluorouracil
.
Clin Cancer Res
2003
;
9
:
4116
24
.
17.
Salonga
D
,
Danenberg
KD
,
Johnson
M
,
Metzger
R
,
Groshen
S
,
Tsao-Wei
DD
, et al
Colorectal tumors responding to 5-fluorouracil have low gene expression levels of dihydropyrimidine dehydrogenase, thymidylate synthase, and thymidine phosphorylase
.
Clin Cancer Res
2000
;
6
:
1322
7
.
18.
Soong
R
,
Shah
N
,
Salto-Tellez
M
,
Tai
BC
,
Soo
RA
,
Han
HC
, et al
Prognostic significance of thymidylate synthase, dihydropyrimidine dehydrogenase and thymidine phosphorylase protein expression in colorectal cancer patients treated with or without 5-fluorouracil-based chemotherapy
.
Ann Oncol
2008
;
19
:
915
9
.
19.
Braun
MS
,
Richman
SD
,
Quirke
P
,
Daly
C
,
Adlard
JW
,
Elliott
F
, et al
Predictive biomarkers of chemotherapy efficacy in colorectal cancer: results from the UK MRC FOCUS trial
.
J Clin Oncol
2008
;
26
:
2690
8
.
20.
Vallböhmer
D
,
Yang
DY
,
Kuramochi
H
,
Shimizu
D
,
Danenberg
KD
,
Lindebjerg
J
, et al
DPD is a molecular determinant of capecitabine efficacy in colorectal cancer
.
Int J Oncol
2007
;
31
:
413
8
.
21.
Koopman
M
,
Venderbosch
S
,
van Tinteren
H
,
Ligtenberg
MJ
,
Nagtegaal
I
,
Van Krieken
JH
, et al
Predictive and prognostic markers for the outcome of chemotherapy in advanced colorectal cancer, a retrospective analysis of the phase III randomised CAIRO study
.
Eur J Cancer
2009
;
45
:
1999
2006
.
22.
Deenen
MJ
,
Tol
J
,
Burylo
AM
,
Doodeman
VD
,
de Boer
A
,
Vincent
A
, et al
Relationship between single nucleotide polymorphisms and haplotypes in DPYD and toxicity and efficacy of capecitabine in advanced colorectal cancer
.
Clin Cancer Res
2011
;
17
:
3455
68
.
23.
Meulendijks
D
,
Henricks
LM
,
Sonke
GS
,
Deenen
MJ
,
Froehlich
TK
,
Amstutz
U
, et al
Clinical relevance of DPYD variants c.1679T>G, c.1236G>A/HapB3, and c.1601G>A as predictors of severe fluoropyrimidine-associated toxicity: a systematic review and meta-analysis of individual patient data
.
Lancet Oncol
2015
;
16
:
1639
50
.
24.
Henricks
LM
,
Lunenburg
CATC
,
de Man
FM
,
Meulendijks
D
,
Frederix
GWJ
,
Kienhuis
E
, et al
DPYD genotype-guided dose individualisation of fluoropyrimidine therapy in patients with cancer: a prospective safety analysis
.
Lancet Oncol
2018
;
19
:
1459
67
.
25.
Cheng
L
,
Li
M
,
Hu
J
,
Ren
W
,
Xie
L
,
Sun
ZP
, et al
UGT1A1*6 polymorphisms are correlated with irinotecan-induced toxicity: a system review and meta-analysis in Asians
.
Cancer Chemother Pharmacol
2014
;
73
:
551
60
.
26.
Tejpar
S
,
Yan
P
,
Piessevaux
H
,
Dietrich
D
,
Brauchli
P
,
Klingbiel
D
, et al
Clinical and pharmacogenetic determinants of 5-fluorouracyl/leucovorin/irinotecan toxicity: Results of the PETACC-3 trial
.
Eur J Cancer
2018
;
99
:
66
77
.
27.
Shirota
Y
,
Stoehlmacher
J
,
Brabender
J
,
Xiong
YP
,
Uetake
H
,
Danenberg
KD
, et al
ERCC1 and thymidylate synthase mRNA levels predict survival for colorectal cancer patients receiving combination oxaliplatin and fluorouracil chemotherapy
.
J Clin Oncol
2001
;
19
:
4298
304
.
28.
Viguier
J
,
Boige
V
,
Miquel
C
,
Pocard
M
,
Giraudeau
B
,
Sabourin
JC
, et al
ERCC1 codon 118 polymorphism is a predictive factor for the tumor response to oxaliplatin/5-fluorouracil combination chemotherapy in patients with advanced colorectal cancer
.
Clin Cancer Res
2005
;
11
:
6212
7
.
29.
Parikh
AR
,
Lee
FC
,
Yau
L
,
Koh
H
,
Knost
J
,
Mitchell
EP
, et al
MAVERICC, a randomized, biomarker-stratified, phase II study of mFOLFOX6-bevacizumab versus FOLFIRI-bevacizumab as first-line chemotherapy in metastatic colorectal cancer
.
Clin Cancer Res
2019
;
25
:
2988
95
.
30.
Formica
V
,
Doldo
E
,
Antonetti
FR
,
Nardecchia
A
,
Ferroni
P
,
Riondino
S
, et al
Biological and predictive role of ERCC1 polymorphisms in cancer
.
Crit Rev Oncol Hematol
2017
;
111
:
133
43
.
31.
Douillard
JY
,
Siena
S
,
Cassidy
J
,
Tabernero
J
,
Burkes
R
,
Barugel
M
, et al
Randomized, phase III trial of panitumumab with infusional fluorouracil, leucovorin, and oxaliplatin (FOLFOX4) versus FOLFOX4 alone as first-line treatment in patients with previously untreated metastatic colorectal cancer: the PRIME study
.
J Clin Oncol
2010
;
28
:
4697
705
.
32.
Karapetis
CS
,
Khambata-Ford
S
,
Jonker
DJ
,
O'Callaghan
CJ
,
Tu
D
,
Tebbutt
NC
, et al
K-ras mutations and benefit from cetuximab in advanced colorectal cancer
.
N Engl J Med
2008
;
359
:
1757
65
.
33.
Douillard
JY
,
Oliner
KS
,
Siena
S
,
Tabernero
J
,
Burkes
R
,
Barugel
M
, et al
Panitumumab-FOLFOX4 treatment and RAS mutations in colorectal cancer
.
N Engl J Med
2013
;
369
:
1023
34
.
34.
De Roock
W
,
Jonker
DJ
,
Di Nicolantonio
F
,
Sartore-Bianchi
A
,
Tu
D
,
Siena
S
, et al
Association of KRAS p.G13D mutation with outcome in patients with chemotherapy-refractory metastatic colorectal cancer treated with cetuximab
.
JAMA
2010
;
304
:
1812
20
.
35.
Tejpar
S
,
Celik
I
,
Schlichting
M
,
Sartorius
U
,
Bokemeyer
C
,
Van Cutsem
E
. 
Association of KRAS G13D tumor mutations with outcome in patients with metastatic colorectal cancer treated with first-line chemotherapy with or without cetuximab
.
J Clin Oncol
2012
;
30
:
3570
7
.
36.
Peeters
M
,
Douillard
JY
,
Van Cutsem
E
,
Siena
S
,
Zhang
K
,
Williams
R
, et al
Mutant KRAS codon 12 and 13 alleles in patients with metastatic colorectal cancer: assessment as prognostic and predictive biomarkers of response to panitumumab
.
J Clin Oncol
2013
;
31
:
759
65
.
37.
Gajate
P
,
Sastre
J
,
Bando
I
,
Alonso
T
,
Cillero
L
,
Sanz
J
, et al
Influence of KRAS p.G13D mutation in patients with metastatic colorectal cancer treated with cetuximab
.
Clin Colorectal Cancer
2012
;
11
:
291
6
.
38.
Rabara
D
,
Tran
TH
,
Dharmaiah
S
,
Stephens
RM
,
McCormick
F
,
Simanshu
DK
, et al
KRAS G13D sensitivity to neurofibromin-mediated GTP hydrolysis
.
Proc Natl Acad Sci U S A
2019
;
116
:
22122
31
.
39.
McFall
T
,
Diedrich
JK
,
Mengistu
M
,
Littlechild
SL
,
Paskvan
KV
,
Sisk-Hackworth
L
, et al
A systems mechanism for KRAS mutant allele-specific responses to targeted therapy
.
Sci Signal
2019
;
12
:
eaaw8288
.
40.
Khambata-Ford
S
,
Garrett
CR
,
Meropol
NJ
,
Basik
M
,
Harbison
CT
,
Wu
S
, et al
Expression of epiregulin and amphiregulin and K-ras mutation status predict disease control in metastatic colorectal cancer patients treated with cetuximab
.
J Clin Oncol
2007
;
25
:
3230
7
.
41.
Jonker
DJ
,
Karapetis
CS
,
Harbison
C
,
O'Callaghan
CJ
,
Tu
D
,
Simes
RJ
, et al
Epiregulin gene expression as a biomarker of benefit from cetuximab in the treatment of advanced colorectal cancer
.
Br J Cancer
2014
;
110
:
648
55
.
42.
Hurwitz
H
,
Fehrenbacher
L
,
Novotny
W
,
Cartwright
T
,
Hainsworth
J
,
Heim
W
, et al
Bevacizumab plus irinotecan, fluorouracil, and leucovorin for metastatic colorectal cancer
.
N Engl J Med
2004
;
350
:
2335
42
.
43.
Saltz
LB
,
Clarke
S
,
Díaz-Rubio
E
,
Scheithauer
W
,
Figer
A
,
Wong
R
, et al
Bevacizumab in combination with oxaliplatin-based chemotherapy as first-line therapy in metastatic colorectal cancer: a randomized phase III study
.
J Clin Oncol
2008
;
26
:
2013
9
.
44.
Heinemann
V
,
von Weikersthal
LF
,
Decker
T
,
Kiani
A
,
Vehling-Kaiser
U
,
Al-Batran
SE
, et al
FOLFIRI plus cetuximab versus FOLFIRI plus bevacizumab as first-line treatment for patients with metastatic colorectal cancer (FIRE-3): a randomised, open-label, phase 3 trial
.
Lancet Oncol
2014
;
15
:
1065
75
.
45.
Van Cutsem
E
,
Tabernero
J
,
Lakomy
R
,
Prenen
H
,
Prausová
J
,
Macarulla
T
, et al
Addition of aflibercept to fluorouracil, leucovorin, and irinotecan improves survival in a phase III randomized trial in patients with metastatic colorectal cancer previously treated with an oxaliplatin-based regimen
.
J Clin Oncol
2012
;
30
:
3499
506
.
46.
Tabernero
J
,
Yoshino
T
,
Cohn
AL
,
Obermannova
R
,
Bodoky
G
,
Garcia-Carbonero
R
, et al
Ramucirumab versus placebo in combination with second-line FOLFIRI in patients with metastatic colorectal carcinoma that progressed during or after first-line therapy with bevacizumab, oxaliplatin, and a fluoropyrimidine (RAISE): a randomised, double-blind, multicentre, phase 3 study
.
Lancet Oncol
2015
;
16
:
499
508
.
47.
Grothey
A
,
Van Cutsem
E
,
Sobrero
A
,
Siena
S
,
Falcone
A
,
Ychou
M
, et al
Regorafenib monotherapy for previously treated metastatic colorectal cancer (CORRECT): an international, multicentre, randomised, placebo-controlled, phase 3 trial
.
Lancet
2013
;
381
:
303
12
.
48.
Jubb
AM
,
Harris
AL
. 
Biomarkers to predict the clinical efficacy of bevacizumab in cancer
.
Lancet Oncol
2010
;
11
:
1172
83
.
49.
Bates
DO
,
Catalano
PJ
,
Symonds
KE
,
Varey
AH
,
Ramani
P
,
O'Dwyer
PJ
, et al
Association between VEGF splice isoforms and progression-free survival in metastatic colorectal cancer patients treated with bevacizumab
.
Clin Cancer Res
2012
;
18
:
6384
91
.
50.
Weickhardt
AJ
,
Williams
DS
,
Lee
CK
,
Chionh
F
,
Simes
J
,
Murone
C
, et al
Vascular endothelial growth factor D expression is a potential biomarker of bevacizumab benefit in colorectal cancer
.
Br J Cancer
2015
;
113
:
37
45
.
51.
Bai
L
,
Wang
F
,
Zhang
DS
,
Li
C
,
Jin
Y
,
Wang
DS
, et al
A plasma cytokine and angiogenic factor (CAF) analysis for selection of bevacizumab therapy in patients with metastatic colorectal cancer
.
Sci Rep
2015
;
5
:
17717
.
52.
Kopetz
S
,
Hoff
PM
,
Morris
JS
,
Wolff
RA
,
Eng
C
,
Glover
KY
, et al
Phase II trial of infusional fluorouracil, irinotecan, and bevacizumab for metastatic colorectal cancer: efficacy and circulating angiogenic biomarkers associated with therapeutic resistance
.
J Clin Oncol
2010
;
28
:
453
9
.
53.
Van Cutsem
E
,
de Haas
S
,
Kang
YK
,
Ohtsu
A
,
Tebbutt
NC
,
Ming Xu
J
, et al
Bevacizumab in combination with chemotherapy as first-line therapy in advanced gastric cancer: a biomarker evaluation from the AVAGAST randomized phase III trial
.
J Clin Oncol
2012
;
30
:
2119
27
.
54.
Schneider
BP
,
Wang
M
,
Radovich
M
,
Sledge
GW
,
Badve
S
,
Thor
A
, et al
Association of vascular endothelial growth factor and vascular endothelial growth factor receptor-2 genetic polymorphisms with outcome in a trial of paclitaxel compared with paclitaxel plus bevacizumab in advanced breast cancer: ECOG 2100
.
J Clin Oncol
2008
;
26
:
4672
8
.
55.
de Haas
S
,
Delmar
P
,
Bansal
AT
,
Moisse
M
,
Miles
DW
,
Leighl
N
, et al
Genetic variability of VEGF pathway genes in six randomized phase III trials assessing the addition of bevacizumab to standard therapy
.
Angiogenesis
2014
;
17
:
909
20
.
56.
Lambrechts
D
,
Claes
B
,
Delmar
P
,
Reumers
J
,
Mazzone
M
,
Yesilyurt
BT
, et al
VEGF pathway genetic variants as biomarkers of treatment outcome with bevacizumab: an analysis of data from the AViTA and AVOREN randomised trials
.
Lancet Oncol
2012
;
13
:
724
33
.
57.
Barat
A
,
Smeets
D
,
Moran
B
,
Zhang
W
,
Cao
S
,
Das
S
, et al
Combination of variations in inflammation- and endoplasmic reticulum-associated genes as putative biomarker for bevacizumab response in KRAS wild-type colorectal cancer
.
Sci Rep
2020
;
10
:
9778
.
58.
van Dijk
E
,
Biesma
HD
,
Cordes
M
,
Smeets
D
,
Neerincx
M
,
Das
S
, et al
Loss of chromosome 18q11.2-q12.1 is predictive for survival in patients with metastatic colorectal cancer treated with bevacizumab
.
J Clin Oncol
2018
;
36
:
2052
60
.
59.
Smeets
D
,
Miller
IS
,
O'Connor
DP
,
Das
S
,
Moran
B
,
Boeckx
B
, et al
Copy number load predicts outcome of metastatic colorectal cancer patients receiving bevacizumab combination therapy
.
Nat Commun
2018
;
9
:
4112
.
60.
Liu
Y
,
Lyu
J
,
Bell Burdett
K
,
Sibley
AB
,
Hatch
AJ
,
Starr
MD
, et al
Prognostic and predictive biomarkers in metastatic colorectal cancer patients receiving regorafenib
.
Mol Cancer Ther
2020
;
19
:
2146
54
.
61.
Suenaga
M
,
Schirripa
M
,
Cao
S
,
Zhang
W
,
Yang
D
,
Ning
Y
, et al
Gene polymorphisms in the CCL5/CCR5 pathway as a genetic biomarker for outcome and hand-foot skin reaction in metastatic colorectal cancer patients treated with regorafenib
.
Clin Colorectal Cancer
2018
;
17
:
e395
e414
.
62.
Sartore-Bianchi
A
,
Trusolino
L
,
Martino
C
,
Bencardino
K
,
Lonardi
S
,
Bergamo
F
, et al
Dual-targeted therapy with trastuzumab and lapatinib in treatment-refractory, KRAS codon 12/13 wild-type, HER2-positive metastatic colorectal cancer (HERACLES): a proof-of-concept, multicentre, open-label, phase 2 trial
.
Lancet Oncol
2016
;
17
:
738
46
.
63.
Meric-Bernstam
F
,
Hurwitz
H
,
Raghav
KPS
,
McWilliams
RR
,
Fakih
M
,
VanderWalde
A
, et al
Pertuzumab plus trastuzumab for HER2-amplified metastatic colorectal cancer (MyPathway): an updated report from a multicentre, open-label, phase 2a, multiple basket study
.
Lancet Oncol
2019
;
20
:
518
30
.
64.
Pietrantonio
F
,
Di Nicolantonio
F
,
Schrock
AB
,
Lee
J
,
Tejpar
S
,
Sartore-Bianchi
A
, et al
ALK, ROS1, and NTRK rearrangements in metastatic colorectal cancer
.
J Natl Cancer Inst
2017
;
109
:
djx089
.
65.
Sartore-Bianchi
A
,
Ardini
E
,
Bosotti
R
,
Amatu
A
,
Valtorta
E
,
Somaschini
A
, et al
Sensitivity to entrectinib associated with a novel LMNA-NTRK1 gene fusion in metastatic colorectal cancer
.
J Natl Cancer Inst
2016
;
108
:
djv306
.
66.
Siravegna
G
,
Sartore-Bianchi
A
,
Mussolin
B
,
Cassingena
A
,
Amatu
A
,
Novara
L
, et al
Tracking a CAD-ALK gene rearrangement in urine and blood of a colorectal cancer patient treated with an ALK inhibitor
.
Ann Oncol
2017
;
28
:
1302
8
.
67.
Li
AY
,
McCusker
MG
,
Russo
A
,
Scilla
KA
,
Gittens
A
,
Arensmeyer
K
, et al
RET fusions in solid tumors
.
Cancer Treat Rev
2019
;
81
:
101911
.
68.
Gozgit
JM
,
Chen
TH
,
Song
Y
,
Wardwell
S
,
Wang
F
,
Cai
J
, et al
RET fusions observed in lung and colorectal cancers are sensitive to ponatinib
.
Oncotarget
2018
;
9
:
29654
64
.
69.
Tol
J
,
Nagtegaal
ID
,
Punt
CJ
. 
BRAF mutation in metastatic colorectal cancer
.
N Engl J Med
2009
;
361
:
98
9
.
70.
De Roock
W
,
De Vriendt
V
,
Normanno
N
,
Ciardiello
F
,
Tejpar
S
. 
KRAS, BRAF, PIK3CA, and PTEN mutations: implications for targeted therapies in metastatic colorectal cancer
.
Lancet Oncol
2011
;
12
:
594
603
.
71.
Ahronian
LG
,
Sennott
EM
,
Van Allen
EM
,
Wagle
N
,
Kwak
EL
,
Faris
JE
, et al
Clinical acquired resistance to RAF inhibitor combinations in BRAF-mutant colorectal cancer through MAPK pathway alterations
.
Cancer Discov
2015
;
5
:
358
67
.
72.
Prahallad
A
,
Sun
C
,
Huang
S
,
Di Nicolantonio
F
,
Salazar
R
,
Zecchin
D
, et al
Unresponsiveness of colon cancer to BRAF(V600E) inhibition through feedback activation of EGFR
.
Nature
2012
;
483
:
100
3
.
73.
Kopetz
S
,
Grothey
A
,
Yaeger
R
,
Van Cutsem
E
,
Desai
J
,
Yoshino
T
, et al
Encorafenib, binimetinib, and cetuximab in BRAF V600E-mutated colorectal cancer
.
N Engl J Med
2019
;
381
:
1632
43
.
74.
Amodio
V
,
Yaeger
R
,
Arcella
P
,
Cancelliere
C
,
Lamba
S
,
Lorenzato
A
, et al
EGFR blockade reverts resistance to KRAS
.
Cancer Discov
2020
;
10
:
1129
39
.
75.
Germano
G
,
Lamba
S
,
Rospo
G
,
Barault
L
,
Magrì
A
,
Maione
F
, et al
Inactivation of DNA repair triggers neoantigen generation and impairs tumour growth
.
Nature
2017
;
552
:
116
20
.
76.
Le
DT
,
Uram
JN
,
Wang
H
,
Bartlett
BR
,
Kemberling
H
,
Eyring
AD
, et al
PD-1 blockade in tumors with mismatch-repair deficiency
.
N Engl J Med
2015
;
372
:
2509
20
.
77.
Overman
MJ
,
McDermott
R
,
Leach
JL
,
Lonardi
S
,
Lenz
HJ
,
Morse
MA
, et al
Nivolumab in patients with metastatic DNA mismatch repair-deficient or microsatellite instability-high colorectal cancer (CheckMate 142): an open-label, multicentre, phase 2 study
.
Lancet Oncol
2017
;
18
:
1182
91
.
78.
Le
DT
,
Durham
JN
,
Smith
KN
,
Wang
H
,
Bartlett
BR
,
Aulakh
LK
, et al
Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade
.
Science
2017
;
357
:
409
13
.
79.
Canon
J
,
Rex
K
,
Saiki
AY
,
Mohr
C
,
Cooke
K
,
Bagal
D
, et al
The clinical KRAS(G12C) inhibitor AMG 510 drives anti-tumour immunity
.
Nature
2019
;
575
:
217
23
.
80.
Catalano
I
,
Grassi
E
,
Bertotti
A
,
Trusolino
L
. 
Immunogenomics of colorectal tumors: facts and hypotheses on an evolving saga
.
Trends Cancer
2019
;
5
:
779
88
.
81.
Hidalgo
M
,
Amant
F
,
Biankin
AV
,
Budinská
E
,
Byrne
AT
,
Caldas
C
, et al
Patient-derived xenograft models: an emerging platform for translational cancer research
.
Cancer Discov
2014
;
4
:
998
1013
.
82.
Gao
H
,
Korn
JM
,
Ferretti
S
,
Monahan
JE
,
Wang
Y
,
Singh
M
, et al
High-throughput screening using patient-derived tumor xenografts to predict clinical trial drug response
.
Nat Med
2015
;
21
:
1318
25
.
83.
Julien
S
,
Merino-Trigo
A
,
Lacroix
L
,
Pocard
M
,
Goéré
D
,
Mariani
P
, et al
Characterization of a large panel of patient-derived tumor xenografts representing the clinical heterogeneity of human colorectal cancer
.
Clin Cancer Res
2012
;
18
:
5314
28
.
84.
Schütte
M
,
Risch
T
,
Abdavi-Azar
N
,
Boehnke
K
,
Schumacher
D
,
Keil
M
, et al
Molecular dissection of colorectal cancer in pre-clinical models identifies biomarkers predicting sensitivity to EGFR inhibitors
.
Nat Commun
2017
;
8
:
14262
.
85.
Bertotti
A
,
Migliardi
G
,
Galimi
F
,
Sassi
F
,
Torti
D
,
Isella
C
, et al
A molecularly annotated platform of patient-derived xenografts (“xenopatients”) identifies HER2 as an effective therapeutic target in cetuximab-resistant colorectal cancer
.
Cancer Discov
2011
;
1
:
508
23
.
86.
Van Cutsem
E
,
Köhne
CH
,
Hitre
E
,
Zaluski
J
,
Chang Chien
CR
,
Makhson
A
, et al
Cetuximab and chemotherapy as initial treatment for metastatic colorectal cancer
.
N Engl J Med
2009
;
360
:
1408
17
.
87.
De Roock
W
,
Claes
B
,
Bernasconi
D
,
De Schutter
J
,
Biesmans
B
,
Fountzilas
G
, et al
Effects of KRAS, BRAF, NRAS, and PIK3CA mutations on the efficacy of cetuximab plus chemotherapy in chemotherapy-refractory metastatic colorectal cancer: a retrospective consortium analysis
.
Lancet Oncol
2010
;
11
:
753
62
.
88.
Guinney
J
,
Dienstmann
R
,
Wang
X
,
de Reyniès
A
,
Schlicker
A
,
Soneson
C
, et al
The consensus molecular subtypes of colorectal cancer
.
Nat Med
2015
;
21
:
1350
6
.
89.
Isella
C
,
Terrasi
A
,
Bellomo
SE
,
Petti
C
,
Galatola
G
,
Muratore
A
, et al
Stromal contribution to the colorectal cancer transcriptome
.
Nat Genet
2015
;
47
:
312
9
.
90.
Isella
C
,
Brundu
F
,
Bellomo
SE
,
Galimi
F
,
Zanella
E
,
Porporato
R
, et al
Selective analysis of cancer-cell intrinsic transcriptional traits defines novel clinically relevant subtypes of colorectal cancer
.
Nat Commun
2017
;
8
:
15107
.
91.
Dunne
PD
,
Alderdice
M
,
O'Reilly
PG
,
Roddy
AC
,
McCorry
AMB
,
Richman
S
, et al
Cancer-cell intrinsic gene expression signatures overcome intratumoural heterogeneity bias in colorectal cancer patient classification
.
Nat Commun
2017
;
8
:
15657
.
92.
Marusyk
A
,
Almendro
V
,
Polyak
K
. 
Intra-tumour heterogeneity: a looking glass for cancer?
Nat Rev Cancer
2012
;
12
:
323
34
.
93.
Kreso
A
,
O'Brien
CA
,
van Galen
P
,
Gan
OI
,
Notta
F
,
Brown
AM
, et al
Variable clonal repopulation dynamics influence chemotherapy response in colorectal cancer
.
Science
2013
;
339
:
543
8
.
94.
Lenos
KJ
,
Miedema
DM
,
Lodestijn
SC
,
Nijman
LE
,
van den Bosch
T
,
Romero Ros
X
, et al
Stem cell functionality is microenvironmentally defined during tumour expansion and therapy response in colon cancer
.
Nat Cell Biol
2018
;
20
:
1193
202
.
95.
van der Heijden
M
,
Miedema
DM
,
Waclaw
B
,
Veenstra
VL
,
Lecca
MC
,
Nijman
LE
, et al
Spatiotemporal regulation of clonogenicity in colorectal cancer xenografts
.
Proc Natl Acad Sci U S A
2019
;
116
:
6140
5
.
96.
Lamprecht
S
,
Schmidt
EM
,
Blaj
C
,
Hermeking
H
,
Jung
A
,
Kirchner
T
, et al
Multicolor lineage tracing reveals clonal architecture and dynamics in colon cancer
.
Nat Commun
2017
;
8
:
1406
.
97.
Leto
SM
,
Sassi
F
,
Catalano
I
,
Torri
V
,
Migliardi
G
,
Zanella
ER
, et al
Sustained inhibition of HER3 and EGFR is necessary to induce regression of HER2-amplified gastrointestinal carcinomas
.
Clin Cancer Res
2015
;
21
:
5519
31
.
98.
Bardelli
A
,
Corso
S
,
Bertotti
A
,
Hobor
S
,
Valtorta
E
,
Siravegna
G
, et al
Amplification of the MET receptor drives resistance to anti-EGFR therapies in colorectal cancer
.
Cancer Discov
2013
;
3
:
658
73
.
99.
Bertotti
A
,
Papp
E
,
Jones
S
,
Adleff
V
,
Anagnostou
V
,
Lupo
B
, et al
The genomic landscape of response to EGFR blockade in colorectal cancer
.
Nature
2015
;
526
:
263
7
.
100.
Kavuri
SM
,
Jain
N
,
Galimi
F
,
Cottino
F
,
Leto
SM
,
Migliardi
G
, et al
HER2 activating mutations are targets for colorectal cancer treatment
.
Cancer Discov
2015
;
5
:
832
41
.
101.
Zanella
ER
,
Galimi
F
,
Sassi
F
,
Migliardi
G
,
Cottino
F
,
Leto
SM
, et al
IGF2 is an actionable target that identifies a distinct subpopulation of colorectal cancer patients with marginal response to anti-EGFR therapies
.
Sci Transl Med
2015
;
7
:
272ra12
.
102.
Russo
M
,
Siravegna
G
,
Blaszkowsky
LS
,
Corti
G
,
Crisafulli
G
,
Ahronian
LG
, et al
Tumor heterogeneity and lesion-specific response to targeted therapy in colorectal cancer
.
Cancer Discov
2016
;
6
:
147
53
.
103.
Bivona
TG
,
Doebele
RC
. 
A framework for understanding and targeting residual disease in oncogene-driven solid cancers
.
Nat Med
2016
;
22
:
472
8
.
104.
Lupo
B
,
Sassi
F
,
Pinnelli
M
,
Galimi
F
,
Zanella
ER
,
Vurchio
V
, et al
Colorectal cancer residual disease at maximal response to EGFR blockade displays a druggable Paneth cell-like phenotype
.
Sci Transl Med
2020
;
12
:
eaax8313
.
105.
Sequist
LV
,
Waltman
BA
,
Dias-Santagata
D
,
Digumarthy
S
,
Turke
AB
,
Fidias
P
, et al
Genotypic and histological evolution of lung cancers acquiring resistance to EGFR inhibitors
.
Sci Transl Med
2011
;
3
:
75ra26
.
106.
Zou
M
,
Toivanen
R
,
Mitrofanova
A
,
Floch
N
,
Hayati
S
,
Sun
Y
, et al
Transdifferentiation as a mechanism of treatment resistance in a mouse model of castration-resistant prostate cancer
.
Cancer Discov
2017
;
7
:
736
49
.
107.
Jackstadt
R
,
Sansom
OJ
. 
Mouse models of intestinal cancer
.
J Pathol
2016
;
238
:
141
51
.
108.
Shibata
H
,
Toyama
K
,
Shioya
H
,
Ito
M
,
Hirota
M
,
Hasegawa
S
, et al
Rapid colorectal adenoma formation initiated by conditional targeting of the Apc gene
.
Science
1997
;
278
:
120
3
.
109.
Takaku
K
,
Oshima
M
,
Miyoshi
H
,
Matsui
M
,
Seldin
MF
,
Taketo
MM
. 
Intestinal tumorigenesis in compound mutant mice of both Dpc4 (Smad4) and Apc genes
.
Cell
1998
;
92
:
645
56
.
110.
Halberg
RB
,
Katzung
DS
,
Hoff
PD
,
Moser
AR
,
Cole
CE
,
Lubet
RA
, et al
Tumorigenesis in the multiple intestinal neoplasia mouse: redundancy of negative regulators and specificity of modifiers
.
Proc Natl Acad Sci U S A
2000
;
97
:
3461
6
.
111.
Hamamoto
T
,
Beppu
H
,
Okada
H
,
Kawabata
M
,
Kitamura
T
,
Miyazono
K
, et al
Compound disruption of smad2 accelerates malignant progression of intestinal tumors in apc knockout mice
.
Cancer Res
2002
;
62
:
5955
61
.
112.
Sansom
OJ
,
Meniel
V
,
Wilkins
JA
,
Cole
AM
,
Oien
KA
,
Marsh
V
, et al
Loss of Apc allows phenotypic manifestation of the transforming properties of an endogenous K-ras oncogene in vivo
.
Proc Natl Acad Sci U S A
2006
;
103
:
14122
7
.
113.
Boutin
AT
,
Liao
WT
,
Wang
M
,
Hwang
SS
,
Karpinets
TV
,
Cheung
H
, et al
Oncogenic Kras drives invasion and maintains metastases in colorectal cancer
.
Genes Dev
2017
;
31
:
370
82
.
114.
Sakai
E
,
Nakayama
M
,
Oshima
H
,
Kouyama
Y
,
Niida
A
,
Fujii
S
, et al
Combined mutation of Apc, Kras, and Tgfbr2 effectively drives metastasis of intestinal cancer
.
Cancer Res
2018
;
78
:
1334
46
.
115.
Tauriello
DVF
,
Palomo-Ponce
S
,
Stork
D
,
Berenguer-Llergo
A
,
Badia-Ramentol
J
,
Iglesias
M
, et al
TGFβ drives immune evasion in genetically reconstituted colon cancer metastasis
.
Nature
2018
;
554
:
538
43
.
116.
Jackstadt
R
,
van Hooff
SR
,
Leach
JD
,
Cortes-Lavaud
X
,
Lohuis
JO
,
Ridgway
RA
, et al
Epithelial NOTCH signaling rewires the tumor microenvironment of colorectal cancer to drive poor-prognosis subtypes and metastasis
.
Cancer Cell
2019
;
36
:
319
36
.
117.
Crystal
AS
,
Shaw
AT
,
Sequist
LV
,
Friboulet
L
,
Niederst
MJ
,
Lockerman
EL
, et al
Patient-derived models of acquired resistance can identify effective drug combinations for cancer
.
Science
2014
;
346
:
1480
6
.
118.
Drost
J
,
Clevers
H
. 
Organoids in cancer research
.
Nat Rev Cancer
2018
;
18
:
407
18
.
119.
Sato
T
,
Vries
RG
,
Snippert
HJ
,
van de Wetering
M
,
Barker
N
,
Stange
DE
, et al
Single Lgr5 stem cells build crypt-villus structures in vitro without a mesenchymal niche
.
Nature
2009
;
459
:
262
5
.
120.
Sato
T
,
Clevers
H
. 
Growing self-organizing mini-guts from a single intestinal stem cell: mechanism and applications
.
Science
2013
;
340
:
1190
4
.
121.
van de Wetering
M
,
Francies
HE
,
Francis
JM
,
Bounova
G
,
Iorio
F
,
Pronk
A
, et al
Prospective derivation of a living organoid biobank of colorectal cancer patients
.
Cell
2015
;
161
:
933
45
.
122.
Fujii
M
,
Shimokawa
M
,
Date
S
,
Takano
A
,
Matano
M
,
Nanki
K
, et al
A colorectal tumor organoid library demonstrates progressive loss of niche factor requirements during tumorigenesis
.
Cell Stem Cell
2016
;
18
:
827
38
.
123.
Fumagalli
A
,
Drost
J
,
Suijkerbuijk
SJ
,
van Boxtel
R
,
de Ligt
J
,
Offerhaus
GJ
, et al
Genetic dissection of colorectal cancer progression by orthotopic transplantation of engineered cancer organoids
.
Proc Natl Acad Sci U S A
2017
;
114
:
E2357
E64
.
124.
de Sousa e Melo
F
,
Kurtova
AV
,
Harnoss
JM
,
Kljavin
N
,
Hoeck
JD
,
Hung
J
, et al
A distinct role for Lgr5 stem cells in primary and metastatic colon cancer
.
Nature
2017
;
543
:
676
80
.
125.
Roper
J
,
Tammela
T
,
Cetinbas
NM
,
Akkad
A
,
Roghanian
A
,
Rickelt
S
, et al
In vivo genome editing and organoid transplantation models of colorectal cancer and metastasis
.
Nat Biotechnol
2017
;
35
:
569
76
.
126.
O'Rourke
KP
,
Loizou
E
,
Livshits
G
,
Schatoff
EM
,
Baslan
T
,
Manchado
E
, et al
Transplantation of engineered organoids enables rapid generation of metastatic mouse models of colorectal cancer
.
Nat Biotechnol
2017
;
35
:
577
82
.
127.
Roper
J
,
Tammela
T
,
Akkad
A
,
Almeqdadi
M
,
Santos
SB
,
Jacks
T
, et al
Colonoscopy-based colorectal cancer modeling in mice with CRISPR-Cas9 genome editing and organoid transplantation
.
Nat Protoc
2018
;
13
:
217
34
.
128.
Matano
M
,
Date
S
,
Shimokawa
M
,
Takano
A
,
Fujii
M
,
Ohta
Y
, et al
Modeling colorectal cancer using CRISPR-Cas9-mediated engineering of human intestinal organoids
.
Nat Med
2015
;
21
:
256
62
.
129.
Drost
J
,
van Jaarsveld
RH
,
Ponsioen
B
,
Zimberlin
C
,
van Boxtel
R
,
Buijs
A
, et al
Sequential cancer mutations in cultured human intestinal stem cells
.
Nature
2015
;
521
:
43
7
.
130.
Shimokawa
M
,
Ohta
Y
,
Nishikori
S
,
Matano
M
,
Takano
A
,
Fujii
M
, et al
Visualization and targeting of LGR5
.
Nature
2017
;
545
:
187
92
.
131.
Roerink
SF
,
Sasaki
N
,
Lee-Six
H
,
Young
MD
,
Alexandrov
LB
,
Behjati
S
, et al
Intra-tumour diversification in colorectal cancer at the single-cell level
.
Nature
2018
;
556
:
457
62
.
132.
Sottoriva
A
,
Kang
H
,
Ma
Z
,
Graham
TA
,
Salomon
MP
,
Zhao
J
, et al
A big bang model of human colorectal tumor growth
.
Nat Genet
2015
;
47
:
209
16
.
133.
Saito
T
,
Niida
A
,
Uchi
R
,
Hirata
H
,
Komatsu
H
,
Sakimura
S
, et al
A temporal shift of the evolutionary principle shaping intratumor heterogeneity in colorectal cancer
.
Nat Commun
2018
;
9
:
2884
.
134.
Vlachogiannis
G
,
Hedayat
S
,
Vatsiou
A
,
Jamin
Y
,
Fernández-Mateos
J
,
Khan
K
, et al
Patient-derived organoids model treatment response of metastatic gastrointestinal cancers
.
Science
2018
;
359
:
920
6
.
135.
Ooft
SN
,
Weeber
F
,
Dijkstra
KK
,
McLean
CM
,
Kaing
S
,
van Werkhoven
E
, et al
Patient-derived organoids can predict response to chemotherapy in metastatic colorectal cancer patients
.
Sci Transl Med
2019
;
11
:
eaay2574
.
136.
Ganesh
K
,
Wu
C
,
O'Rourke
KP
,
Szeglin
BC
,
Zheng
Y
,
Sauvé
CG
, et al
A rectal cancer organoid platform to study individual responses to chemoradiation
.
Nat Med
2019
;
25
:
1607
14
.
137.
Yao
Y
,
Xu
X
,
Yang
L
,
Zhu
J
,
Wan
J
,
Shen
L
, et al
Patient-derived organoids predict chemoradiation responses of locally advanced rectal cancer
.
Cell Stem Cell
2020
;
26
:
17
26
.
138.
Ben-David
U
,
Ha
G
,
Tseng
YY
,
Greenwald
NF
,
Oh
C
,
Shih
J
, et al
Patient-derived xenografts undergo mouse-specific tumor evolution
.
Nat Genet
2017
;
49
:
1567
75
.
139.
Woo
XY
,
Giordano
J
,
Srivastava
A
,
Zhao
ZM
,
Lloyd
M
,
de Bruijn
R
, et al
Conservation of copy number profiles during engraftment and passaging of patient-derived cancer xenografts
.
Nat. Genet
2021
;
53
:
86
99
.
140.
Eirew
P
,
Steif
A
,
Khattra
J
,
Ha
G
,
Yap
D
,
Farahani
H
, et al
Dynamics of genomic clones in breast cancer patient xenografts at single-cell resolution
.
Nature
2015
;
518
:
422
6
.
141.
Cho
YB
,
Hong
HK
,
Choi
YL
,
Oh
E
,
Joo
KM
,
Jin
J
, et al
Colorectal cancer patient-derived xenografted tumors maintain characteristic features of the original tumors
.
J Surg Res
2014
;
187
:
502
9
.
142.
Weeber
F
,
van de Wetering
M
,
Hoogstraat
M
,
Dijkstra
KK
,
Krijgsman
O
,
Kuilman
T
, et al
Preserved genetic diversity in organoids cultured from biopsies of human colorectal cancer metastases
.
Proc Natl Acad Sci U S A
2015
;
112
:
13308
11
.
143.
Bolhaqueiro
ACF
,
Ponsioen
B
,
Bakker
B
,
Klaasen
SJ
,
Kucukkose
E
,
van Jaarsveld
RH
, et al
Ongoing chromosomal instability and karyotype evolution in human colorectal cancer organoids
.
Nat Genet
2019
;
51
:
824
34
.
144.
Maekawa
H
,
Miyoshi
H
,
Yamaura
T
,
Itatani
Y
,
Kawada
K
,
Sakai
Y
, et al
A chemosensitivity study of colorectal cancer using xenografts of patient-derived tumor-initiating cells
.
Mol Cancer Ther
2018
;
17
:
2187
96
.
145.
Corcoran
RB
,
Atreya
CE
,
Falchook
GS
,
Kwak
EL
,
Ryan
DP
,
Bendell
JC
, et al
Combined BRAF and MEK inhibition with dabrafenib and trametinib in BRAF V600-mutant colorectal cancer
.
J Clin Oncol
2015
;
33
:
4023
31
.
146.
Clohessy
JG
,
Pandolfi
PP
. 
Mouse hospital and co-clinical trial project–from bench to bedside
.
Nat Rev Clin Oncol
2015
;
12
:
491
8
.
147.
West
MA
,
Roman
A
,
Sayan
E
,
Primrose
JN
,
Wedge
SR
,
Underwood
TJ
, et al
A minimum core outcome dataset for the reporting of preclinical chemotherapeutic drug studies: lessons learned from multiple discordant methodologies in the setting of colorectal cancer
.
Crit Rev Oncol Hematol
2017
;
112
:
80
102
.
148.
Blunt
T
,
Gell
D
,
Fox
M
,
Taccioli
GE
,
Lehmann
AR
,
Jackson
SP
, et al
Identification of a nonsense mutation in the carboxyl-terminal region of DNA-dependent protein kinase catalytic subunit in the scid mouse
.
Proc Natl Acad Sci U S A
1996
;
93
:
10285
90
.
149.
Zitvogel
L
,
Pitt
JM
,
Daillère
R
,
Smyth
MJ
,
Kroemer
G
. 
Mouse models in oncoimmunology
.
Nat Rev Cancer
2016
;
16
:
759
73
.
150.
Sanmamed
MF
,
Rodriguez
I
,
Schalper
KA
,
Oñate
C
,
Azpilikueta
A
,
Rodriguez-Ruiz
ME
, et al
Nivolumab and urelumab enhance antitumor activity of human T lymphocytes engrafted in Rag2-/-IL2Rγnull immunodeficient mice
.
Cancer Res
2015
;
75
:
3466
78
.
151.
Capasso
A
,
Lang
J
,
Pitts
TM
,
Jordan
KR
,
Lieu
CH
,
Davis
SL
, et al
Characterization of immune responses to anti-PD-1 mono and combination immunotherapy in hematopoietic humanized mice implanted with tumor xenografts
.
J Immunother Cancer
2019
;
7
:
37
.
152.
Guichelaar
T
,
Emmelot
ME
,
Rozemuller
H
,
Martini
B
,
Groen
RW
,
Storm
G
, et al
Human regulatory T cells do not suppress the antitumor immunity in the bone marrow: a role for bone marrow stromal cells in neutralizing regulatory T cells
.
Clin Cancer Res
2013
;
19
:
1467
75
.
153.
King
MA
,
Covassin
L
,
Brehm
MA
,
Racki
W
,
Pearson
T
,
Leif
J
, et al
Human peripheral blood leucocyte non-obese diabetic-severe combined immunodeficiency interleukin-2 receptor gamma chain gene mouse model of xenogeneic graft-versus-host-like disease and the role of host major histocompatibility complex
.
Clin Exp Immunol
2009
;
157
:
104
18
.
154.
Bar-Ephraim
YE
,
Kretzschmar
K
,
Clevers
H
. 
Organoids in immunological research
.
Nat Rev Immunol
2020
;
20
:
279
93
.
155.
Dijkstra
KK
,
Cattaneo
CM
,
Weeber
F
,
Chalabi
M
,
van de Haar
J
,
Fanchi
LF
, et al
Generation of tumor-reactive T cells by co-culture of peripheral blood lymphocytes and tumor organoids
.
Cell
2018
;
174
:
1586
98
.
156.
Neal
JT
,
Li
X
,
Zhu
J
,
Giangarra
V
,
Grzeskowiak
CL
,
Ju
J
, et al
Organoid modeling of the tumor immune microenvironment
.
Cell
2018
;
175
:
1972
88
.
157.
Carvalho
MR
,
Barata
D
,
Teixeira
LM
,
Giselbrecht
S
,
Reis
RL
,
Oliveira
JM
, et al
Colorectal tumor-on-a-chip system: a 3D tool for precision onco-nanomedicine
.
Sci Adv
2019
;
5
:
eaaw1317
.