16
REVIEW Open Access Applications of patient-derived tumor xenograft models and tumor organoids Go J. Yoshida 1,2 Abstract Patient-derived tumor xenografts (PDXs), in which tumor fragments surgically dissected from cancer patients are directly transplanted into immunodeficient mice, have emerged as a useful model for translational research aimed at facilitating precision medicine. PDX susceptibility to anti-cancer drugs is closely correlated with clinical data in patients, from whom PDX models have been derived. Accumulating evidence suggests that PDX models are highly effective in predicting the efficacy of both conventional and novel anti-cancer therapeutics. This also allows co- clinical trials,in which pre-clinical investigations in vivo and clinical trials could be performed in parallel or sequentially to assess drug efficacy in patients and PDXs. However, tumor heterogeneity present in PDX models and in the original tumor samples constitutes an obstacle for application of PDX models. Moreover, human stromal cells originally present in tumors dissected from patients are gradually replaced by host stromal cells as the xenograft grows. This replacement by murine stroma could preclude analysis of human tumor-stroma interactions, as some mouse stromal cytokines might not affect human carcinoma cells in PDX models. The present review highlights the biological and clinical significance of PDX models and three-dimensional patient-derived tumor organoid cultures of several kinds of solid tumors, such as those of the colon, pancreas, brain, breast, lung, skin, and ovary. Keywords: Acquired resistance, Avatar models, Carcinoma-associated fibroblasts, Co-clinical trials, Heterogeneity, Immunodeficient mice, Organoids, PDX models, Translational research, Tumor microenvironment Background Since the first investigations to develop drugs using ani- mal models of leukemia were reported in 1950 [1], many types of murine models transplanted with human tu- mors have been developed to predict responses to chemotherapy. Many human tumor models were gener- ated in immunodeficient mice by subcutaneous or orthotopic injection of tumor cell lines that had been propagated in culture for several months to several de- cades. Although this model served as a gold standard, primarily because of ease of manipulation, tumor cell lines frequently acquired unanticipated phenotypes dur- ing adaptation to in vitro culture conditions that often differed between laboratories, resulting in minimal re- semblance to the parental tumors. For example, immor- talized cells cultured long-term in vitro exhibit changes in tumor hallmarks caused by genetic and epigenetic al- terations. Thus, substantial limitations have precluded the application of conventional xenograft models for drug screening and estimating the pre-clinical efficacy of drugs [2]. To develop a model more likely to mimic human tu- mors, patient-derived tumor xenografts (PDXs) have been established as a useful tool for translational re- search [36]. Implantation of small pieces of tumors sur- gically dissected from cancer patients into highly immunodeficient mice allows tumor growth and subse- quent transplantation into secondary recipient mice. PDXs often maintain the cellular and histopathological structures of the original tumors (Table 1) [79]. Fur- thermore, cytogenetic analysis of tumor cells from PDXs has revealed significant preservation of the genomic and gene expression profiles between PDXs and parental pa- tient tumors [1014]. Notably, sensitivity to standard chemotherapeutics in PDXs closely correlates with clin- ical data in patients from which the PDXs are derived © The Author(s). 2020 Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated. Correspondence: [email protected] 1 Department of Pathology and Oncology, Juntendo University School of Medicine, 2-1-1, Hongo, Bunkyo-ku, Tokyo 113-8412, Japan 2 Department of Immunological Diagnosis, Juntendo University Graduate School of Medicine, 2-1-1, Hongo, Bunkyo-ku, Tokyo 113-8412, Japan Yoshida Journal of Hematology & Oncology (2020) 13:4 https://doi.org/10.1186/s13045-019-0829-z

Applications of patient-derived tumor xenograft models and tumor … · 2020-01-07 · REVIEW Open Access Applications of patient-derived tumor xenograft models and tumor organoids

  • Upload
    others

  • View
    1

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Applications of patient-derived tumor xenograft models and tumor … · 2020-01-07 · REVIEW Open Access Applications of patient-derived tumor xenograft models and tumor organoids

REVIEW Open Access

Applications of patient-derived tumorxenograft models and tumor organoidsGo J. Yoshida1,2

Abstract

Patient-derived tumor xenografts (PDXs), in which tumor fragments surgically dissected from cancer patients aredirectly transplanted into immunodeficient mice, have emerged as a useful model for translational research aimedat facilitating precision medicine. PDX susceptibility to anti-cancer drugs is closely correlated with clinical data inpatients, from whom PDX models have been derived. Accumulating evidence suggests that PDX models are highlyeffective in predicting the efficacy of both conventional and novel anti-cancer therapeutics. This also allows “co-clinical trials,” in which pre-clinical investigations in vivo and clinical trials could be performed in parallel orsequentially to assess drug efficacy in patients and PDXs. However, tumor heterogeneity present in PDX modelsand in the original tumor samples constitutes an obstacle for application of PDX models. Moreover, human stromalcells originally present in tumors dissected from patients are gradually replaced by host stromal cells as thexenograft grows. This replacement by murine stroma could preclude analysis of human tumor-stroma interactions,as some mouse stromal cytokines might not affect human carcinoma cells in PDX models. The present reviewhighlights the biological and clinical significance of PDX models and three-dimensional patient-derived tumororganoid cultures of several kinds of solid tumors, such as those of the colon, pancreas, brain, breast, lung, skin, andovary.

Keywords: Acquired resistance, Avatar models, Carcinoma-associated fibroblasts, Co-clinical trials, Heterogeneity,Immunodeficient mice, Organoids, PDX models, Translational research, Tumor microenvironment

BackgroundSince the first investigations to develop drugs using ani-mal models of leukemia were reported in 1950 [1], manytypes of murine models transplanted with human tu-mors have been developed to predict responses tochemotherapy. Many human tumor models were gener-ated in immunodeficient mice by subcutaneous ororthotopic injection of tumor cell lines that had beenpropagated in culture for several months to several de-cades. Although this model served as a gold standard,primarily because of ease of manipulation, tumor celllines frequently acquired unanticipated phenotypes dur-ing adaptation to in vitro culture conditions that oftendiffered between laboratories, resulting in minimal re-semblance to the parental tumors. For example, immor-talized cells cultured long-term in vitro exhibit changes

in tumor hallmarks caused by genetic and epigenetic al-terations. Thus, substantial limitations have precludedthe application of conventional xenograft models fordrug screening and estimating the pre-clinical efficacy ofdrugs [2].To develop a model more likely to mimic human tu-

mors, patient-derived tumor xenografts (PDXs) havebeen established as a useful tool for translational re-search [3–6]. Implantation of small pieces of tumors sur-gically dissected from cancer patients into highlyimmunodeficient mice allows tumor growth and subse-quent transplantation into secondary recipient mice.PDXs often maintain the cellular and histopathologicalstructures of the original tumors (Table 1) [7–9]. Fur-thermore, cytogenetic analysis of tumor cells from PDXshas revealed significant preservation of the genomic andgene expression profiles between PDXs and parental pa-tient tumors [10–14]. Notably, sensitivity to standardchemotherapeutics in PDXs closely correlates with clin-ical data in patients from which the PDXs are derived

© The Author(s). 2020 Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, andreproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link tothe Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver(http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Correspondence: [email protected] of Pathology and Oncology, Juntendo University School ofMedicine, 2-1-1, Hongo, Bunkyo-ku, Tokyo 113-8412, Japan2Department of Immunological Diagnosis, Juntendo University GraduateSchool of Medicine, 2-1-1, Hongo, Bunkyo-ku, Tokyo 113-8412, Japan

Yoshida Journal of Hematology & Oncology (2020) 13:4 https://doi.org/10.1186/s13045-019-0829-z

Page 2: Applications of patient-derived tumor xenograft models and tumor … · 2020-01-07 · REVIEW Open Access Applications of patient-derived tumor xenograft models and tumor organoids

[12, 13, 15–18]. All of these characteristics demonstratethat PDXs are more useful as predictive experimentalmodels of therapeutic responses.Tumor organoids are generated by three-dimensional

culture of primary cancer cells and have a high successrate [19–25]. Organoid cultures closely recapitulate themorphological and genetic/epigenetic features of theparent tumors (Table 1) [20, 21, 25, 26]. Accumulatingevidence reveals that primary cancer cells subjected tothree-dimensional culture give rise to many differenttypes of primary organoids, in which the heterogeneouscomposition of the original tumors is largely conserved[22, 27, 28]. This culture method provides promising op-portunities to establish large biobanks with relevant clin-ical materials that can be used to perform drugscreening and facilitate chemical discovery [24, 29].Tumor organoids are superior to conventional models,as organoids constitute a minute incarnation of anin vivo organ, and thus can better recapitulate the char-acteristics of the parental tumor, even after many pas-sages. Organoids have enormous potential for theidentification of optimal treatment strategies in individ-ual patients [28].

PDX models maintain in vivo structureA major advantage of PDX models in cancer research isthe retention of the original tumor architecture. Al-though cancer cell lines are frequently transplanted intoimmunocompromised mice as a reproducible method toestablish tumor tissues in vivo, the resultant tumors donot fully exhibit the distinct histopathological character-istics observed in clinical settings. One important con-sideration in this regard is that cells within tissues aresurrounded by an extracellular matrix (ECM), a mesh-work of proteins and proteoglycans consisting of lam-inin, collagen, and fibronectin, which regulates theintegrity of epithelial structure formations. The ECMprovides structural support, stability, flexibility, andshape to the tissue, and also mediates cell polarity, intra-cellular signaling, and cell migration [30, 31]. Intracellu-larly, ECM-induced signaling pathways are transmittedmainly through integrin molecules, which are heterodi-meric transmembrane receptors that mediate cell adhe-sion to the ECM. Integrins act as bridges between the

ECM and the internal cytoskeleton by transducing keyintracellular signals through association with clusters ofkinases and adaptor proteins in focal adhesion com-plexes [32]. These interactions are distinctly mediated byspecific integrin heterodimer binding to individual ECMcomponents [33]. The signals are transmitted via thecytoskeleton to nuclear transcription factors and ultim-ately change the gene expression profile [34]. As such,highly specific and localized signaling cascades can beactivated by the ECM in association with various avail-able growth factors through sequences of reactions in-volving networks of proteases and sulfatases [35, 36].The tumor microenvironment comprises the ECM and

stromal cells, including endothelial cells, pericytes,carcinoma-associated fibroblasts (CAFs), immune cells,and proinflammatory cells [37, 38]. One of the mostprominent cell types in the tumor stroma is CAFs, whichare activated fibroblasts. CAFs often exhibit an α-smooth muscle actin (α-SMA)-positive myofibroblasticphenotype, which is typical of fibroses and wound heal-ing, and is a pro-inflammatory phenotype [36, 39].Tumor-promoting CAFs influence tumor hallmarks topromote cancer growth, progression, and metastaticspread as well as remodeling of the ECM [36, 39, 40].CAFs contribute to ECM remodeling by secreting TGF-β1, which is synthesized as an inactive multidomaincomplex, and is activated through multiple extracellularmechanisms that require integrins, proteases, andthrombospondin-1 [41, 42]. TGF-β1 has a potential rolein the regulation of ECM remodeling during cancer pro-gression. Exposing CAFs to TGF-β promotes anisotropicfiber organization and increased matrix stiffness throughincreased α-SMA expression and RhoA activation [43].These changes in cellular contractility result in the CAF-mediated changes in the ECM architecture.Tumors in the PDX model have similar pathohistologi-

cal and genetic characteristics to the parent tumor, andexhibit similar susceptibility to anti-cancer therapies [7,10, 11, 15, 16]. Therefore, PDX models based on graftingcancer tissue subcutaneously, orthotopically, or under kid-ney capsules in immunodeficient mice provide relevantpre-clinical cancer models. The mouse strains widely usedfor tumor formation and propagation are classified into (i)nude mice, which lack a thymus and are unable to pro-duce T cells; (ii) non-obese diabetic severe combined im-munodeficiency (NOD-SCID) and SCID-beige mice,which lack T and B cells; and (iii) NOD-SCID IL2R-γ null(NSG or NOG) mice, in which T, B, and NK cell activityis completely absent. Due to differential immunologicalimpairments in these models, it is expected that more per-missive mouse strains such as NOD-SCID, SCID, andNSG, will increase the efficiency of xenotransplantationover that of nude mice. In fact, a very low engraftmentsuccess rate (10–25%) was reported after transplanting

Table 1 Comparison between PDX models and organoids

PDX Organoids

Genetic/epigenetic alterations Similar Similar

Pathohistological characteristics Similar Similar

Response to anti-cancer drugs Similar Similar

Use of immunodeficient animals Yes No

Reliability as pre-clinical models Yes Yes

Quantity of cells for establishment Large Small

Yoshida Journal of Hematology & Oncology (2020) 13:4 Page 2 of 16

Page 3: Applications of patient-derived tumor xenograft models and tumor … · 2020-01-07 · REVIEW Open Access Applications of patient-derived tumor xenograft models and tumor organoids

tumor fragments of different pathohistological types intonude mice, whereas transplantation into NOD-SCID miceresulted in a higher engraftment rate (25-40%) for breastcancer, non-small cell lung cancer and malignant melan-oma [44]. An extremely novel study evaluated the use ofzebrafish for PDX, and reported that transparent zebrafishlarvae allow for visualization of single tumor cells andtheir response to treatment, thereby providing a rapidscreening platform [17].However, it is important to note that human stromal

cells are gradually replaced by murine counterparts aftertransplantation into immunodeficient mice, which sug-gests that implanted human cancer cells retain the poten-tial to recruit murine stromal cells to their niche.Importantly, there are some differences between ligandssecreted by human and murine fibroblasts. Human-derived interleukin (IL)-2 stimulates efficiently the prolif-eration of murine T cells, whereas mouse IL-2 stimulateshuman T cells with significantly lower efficiency [45, 46].Furthermore, the T cell stimulating potential of IL-4 ap-pears to be species-specific [45]. On the other hand, IL-15binds to human or mouse IL-15 receptor α with equallyhigh affinity [47], suggesting that human-derived IL-15can function on murine cells. However, human NK cellsare weakly sensitive to murine IL-15 [48]. In order to re-solve the issues related to species specificity, co-implantation of human CAFs and tumor cell suspensionsextracted from PDXs into secondary recipient mice couldprovide an optimal setting for evaluating human tumorcell-stroma cell interactions.

Patient-derived tumor cell organoid culture mimicparental tumorsFrom a histopathological perspective, PDX modelslargely retain parent tumor architecture, as describedabove. Further, at the cellular level, both inter-tumoraland intra-tumoral heterogeneity, as well as the pheno-typic and molecular characteristics of the original cancerare also preserved in PDX models [12, 49, 50]. From thisperspective, human cancer tissues or tumor cells derivedfrom PDX models that mimic the biological and molecu-lar characteristics of the original cancer can be employedto provide clinically relevant donor cells for in vitromodeling. Tumor tissues from PDX models can be usedto generate three-dimensional tissue explants in vitro orprimary cell cultures on a petri dish. Use of PDXs hasthe advantage that the original tissue can be seriallypropagated in vivo, making additional materials availablefor repeated experiments and alleviating the limitationsinherent to collecting multiple patient samples. To morefully represent the range of biological and molecularcharacteristics of clinical samples, a panel of establishedxenograft lines covering multiple cancer tissues of originand subtypes is required for selection of the most

appropriate model to address particular experimentalquestions.Furthermore, mounting evidence suggests that orga-

noids are three-dimensional constructs comprised ofmultiple cell types that originate from pluripotent stemcells by means of self-organization, and are capable ofsimulating the architecture and functionality of nativetissues and organs [25, 51]. Organoids permit bothin vitro and in vivo investigations, and represent one ofthe latest innovations in development of models to re-capitulate the physiologic processes of whole organisms.Organoids have been successfully generated from pri-mary tumors of the breast, colon, pancreas, and prostate[23, 25]. These tumor-derived organoids have emergedas pre-clinical models that have the potential to predictpersonalized response to treatment. For example, a livingbiobank of tumor organoids from patients with meta-static gastrointestinal cancer was demonstrated to recap-itulate the therapeutic responses of these patients toanti-cancer agents in clinical trials [29].

Use of PDX models for precision medicineMounting evidence demonstrates that PDX models havethe potential to predict effectively the efficacy of bothconventional and novel anti-cancer therapeutics, sug-gesting that these models could be employed in “co-clin-ical trials” [52]. Thus, investigations in vivo and inclinical trials could be performed in parallel in order toidentify therapeutic target molecules, even in rare typesof cancer. Particularly, the concept of “co-clinical trials”incorporates patient selection strategies based on mo-lecular abnormalities or the identification of the machin-eries of resistance to anti-cancer agents, for thedevelopment of precision medicine aimed at personaliz-ing anti-cancer treatment. In this context, PDX modelscan be also used as an “avatar model [53],” in whichPDX models obtained from cancer patients enrolled in aclinical trial can be treated with the same therapy ad-ministered to the patient, thereby permitting identifica-tion of novel biomarkers of sensitivity or resistance tothe anti-cancer treatment(s) of interest (Fig. 1).

Colorectal adenocarcinomaAcquired resistance of tumors to chemotherapeutics is amajor reason for treatment failure [54, 55], and the intro-duction of novel drugs or combinational therapy permitsselection of effective therapeutic strategies for second-linetreatment. Based on this concept, Misale et al. treated ad-vanced colorectal adenocarcinoma (CRC) patients withanti-EGFR antibodies as single agents after the initial re-sponse to EGFR inhibitors resulted in disease relapse dueto emerging resistance [12]. Thus, Misale et al. investi-gated how acquisition of resistance to EGFR-targetedtherapies can be reduced in CRC tumor cells using the

Yoshida Journal of Hematology & Oncology (2020) 13:4 Page 3 of 16

Page 4: Applications of patient-derived tumor xenograft models and tumor … · 2020-01-07 · REVIEW Open Access Applications of patient-derived tumor xenograft models and tumor organoids

PDX model. In vitro genetic screening and functional in-vestigations identified that dual blockade of EGFR andMEK prevents acquired resistance, and Misale et al. per-formed experiments in vivo using PDXs derived from aCRC patient carrying a quadruple wild-type gene profile(BRAF, KRAS, NRAS, and PIK3CA), which recapitulatesthe expression profile of patients sensitive to anti-EGFRantibodies. While treatment of PDX models with theMEK inhibitor pimasertib alone only slightly reducedtumor growth, treatment with the EGFR inhibitor cetuxi-mab effectively reduced cancer proliferation by more than70% [12]. Notably, the subsequent regrowth of these tu-mors suggested resistance to drug re-challenge, which issimilar to clinical findings. By contrast, combination treat-ment with cetuximab and pimasertib induced a completeresponse, in which tumor tissues remained undetectablefor more than 6 months. These findings suggested thatcombination treatment is highly likely to inhibit develop-ment of resistant tumors with intra-tumoral heterogeneity,and highlighted the utility of PDX models in establishinghighly effective treatment regimens.Okazawa et al. established green fluorescent protein

(GFP)-labelled CRC PDX-derived organoids to detect

spontaneous micrometastatic lesions [56]. Micrometastases,which are cancer cell depositions smaller than 2 mm, haveoften been overlooked in the pathohistological analysis ofsections from affected distant organs in experimental mur-ine models, because they may not be detected with this ap-proach. Therefore, Okazawa et al. developed a protocol toefficiently transduce GFP lentivirus into PDX-derived CRCorganoids in three-dimensional culture prior to transplant-ation, enabling highly sensitive detection of micrometas-tases in distant organs such as the liver and lungs (Fig. 2).Using this technology, Okazawa et al. employed a PDXmodel to demonstrate that lung micrometastases could bedetected in the majority of engrafted mice 3 months aftertransplantation. Moreover, liver metastases were detectedwithin 1 month after injection of organoids into the spleen.Further, the injection of CRC organoids into the rectal sub-mucosa of immunocompromised mice resulted in meta-static dissemination into the lungs, but not into the liver,likely through the inferior vena cava.

Pancreatic cancerAlthough the progression of pancreatic ductal adenocar-cinoma (PDAC) is driven by constitutive activation of

Fig. 1 Identification of optimal therapeutics using PDX mouse clinical trials. PDX models are potentially useful when the optimal course oftreatment cannot be readily determined for individual patients. For instance, in the illustration, there are three patients (A-C) with gastric cancer,who hope to receive treatment with the novel therapy drug X if its therapeutic efficacy is proven. In this case, it would be time-consuming andrequire significant clinical risk to compare the therapeutic response to conventional drugs and the new drug X without “co-clinical trials.” Whilexenografts derived from patient A respond to drug X, xenografts derived from patient C respond to conventional treatments, but not drug X(step 1). Contrastingly, patient B–derived xenografts partially respond to both therapeutics. This pre-clinical screening by an avatar model ishelpful to determine which treatment would have the optimal outcome in each patient (step 2)

Yoshida Journal of Hematology & Oncology (2020) 13:4 Page 4 of 16

Page 5: Applications of patient-derived tumor xenograft models and tumor … · 2020-01-07 · REVIEW Open Access Applications of patient-derived tumor xenograft models and tumor organoids

RAS/RAF/MEK/ERK signaling, MEK1/2 inhibition withtrametinib is not clinically effective in PDAC patients. Arecent study revealed that trametinib-induced autophagyflux increases the survival of PDAC cells subjected toMEK1/2 inhibition [57]. Combination treatment withtrametinib and chloroquine increased apoptotic celldeath in a PDX model derived from a distant neck me-tastasis that was refractory to the conventional FOLFOXregimen. It is widely accepted that chloroquine obstructsautophagy flux, increasing ubiquitination, p62/SQSTM1activation, and LC3-II accumulation [58]. Furthermore,the growth of orthotopic and/or subcutaneous PDX tu-mors in NOD/SCID mice was synergistically inhibitedby combination treatment with trametinib and chloro-quine [57]. The efficacy of this modality was also dem-onstrated in PDX models using gain-of-functionalmutated NRAS-driven malignant melanoma andBRAF(V600E)-driven colorectal cancer. Notably, thetherapeutic efficacy of this combination was superior tothat of conventional anti-PDAC drugs such as gemcita-bine. The compensatory activation of protective autoph-agy through the ULK1/AMPK/LKB1 axis appears to beresponsible for susceptibility to trametinib in the pres-ence of chloroquine.

Brain tumorsSingh et al. reported that SOX2 expression is markedly el-evated in PDX derived from glioblastoma multiforme(GBM) [59]. They established PDX models derived from20 GBM patients, comprising different subtypes of

putative GBM driver oncogenes such as EGFR, MET, andPDGFRA. Remarkably, all four IDH1 mutant PDX linesalso exhibited high expression levels of SOX2. High SOX2expression was present in GBM PDX that were driven bydifferent oncogenes, suggesting that multiple oncogenicsignaling pathways are likely to converge to drive expres-sion of this pluripotency transcription factor. While PDXtumors maintained the putative oncogenic mutations ofseveral receptor tyrosine kinases (RTKs) in the parentaltumors, the significant increase in SOX2-expressing GBMcells may reflect the functional state of the subset of GBMcells capable of driving tumor growth in the mouse brainrather than genetic differences between the original pa-tient tumor and PDX model. Singh et al. demonstratedthat the transcriptional regulatory network comprised ofSOX2, OLIG2, and ZEB1 is independent of upstreamRTKs, and is capable of driving glioma initiating cells.Mithramycin is an antibiotic isolated from Streptomycesplicatus that binds to specific DNA regions to inhibit tran-scription of specific genes. Notably, mithramycin down-regulated SOX2 and ZEB1 in sarcoma cells [60], andeffectively inhibited growth of a SOX2-positive cell popu-lation that propagates medulloblastoma [61]. Administrat-ing mithramycin in vivo markedly reduced expression ofSOX2, OLIG2, and ZEB1, which coincided with dramaticreductions in tumor growth [59]. Taken together, thesestudies employing PDX models strongly suggest the im-portance evaluating the efficacy of combining mithramy-cin treatment with chemotherapy and radiation therapy infuture investigations.

Fig. 2 High-sensitivity detection of distant micrometastases of PDX-derived organoids by GFP transduction. After a primary colorectaladenocarcinoma (CRC) diagnosed as a moderately differentiated type was surgically resected, CRC cells were subcutaneously implanted into NOGmice to establish a PDX model. PDX tissue was treated with collagenase to obtain tumor cell suspension. CRC organoids were then establishedfrom PDX tissue and expanded in three-dimensional culture using the artificial extracellular matrix after infection with GFP lentivirus. These GFP-labelled organoids implanted orthotopically revealed distant micrometastases in the lungs within 3 months [56]

Yoshida Journal of Hematology & Oncology (2020) 13:4 Page 5 of 16

Page 6: Applications of patient-derived tumor xenograft models and tumor … · 2020-01-07 · REVIEW Open Access Applications of patient-derived tumor xenograft models and tumor organoids

Breast carcinomaPDX models maintain the essential properties of the ori-ginal patient tumors, including metastatic tropism, sug-gesting their physiological relevance for study of humancancer metastasis [4]. In immunodeficient mice, PDXsspontaneously metastasize many of the same organs af-fected in the original patient. In addition, mesenchymalstem cells (MSCs) in the PDX model enhance tumorgrowth rates by promoting angiogenesis, decreasing ne-crosis, and increasing blood volume, which would con-tribute to the observed increase in tumor growth.Lawason et al. demonstrated using the PDX model thatprogression to high metastatic burden is associated withincreased proliferation and Myc expression, which canbe attenuated by the treatment with cyclin-dependentkinase (CDK) inhibitors [8]. In this study, the mostmetastatic PDX had the highest percentage of cancerstem-like basal primary tumor cells, while the leastmetastatic PDX had the lowest. This suggests that pri-mary tumors contain a rare subpopulation of stem-likecells, and that the relative abundance of these cells couldcorrelate with metastatic potential. Thus, Lawason et al.used PDX models to propose a hierarchical model formetastasis, in which metastases are initiated by cancerstem-like cells, which proliferate and differentiate to pro-duce advanced metastatic disease.

Lung cancerChen et al. recently demonstrated an unexpected plasti-city and interaction of lung squamous cancer cells(LSCCs) with the tumor microenvironment [62]. Over-expression of SOX2 in the TUM622 cell line, which wasestablished from a PDX model, enhances spheroid-forming potential and drives a hyperplastic to dysplasticalteration in acinar phenotype, in which apical-basal cellpolarity is disrupted, and solid non-invasive spheroidsare formed. Remarkably, the presence of CAFs inhibitsSOX2-induced dysplasia and restores an acinar-likephenotype, but TUM622 cells appear to exhibitepithelial-mesenchymal transition (EMT) at the invasivefront towards CAFs, thereby forming “teardrop”-shapedstructures [62, 63]. Indeed, CAF-secreted stromal cell-derived factor-1 (SDF-1) promoted EMT and the acqui-sition of stemness in LSCCs [64]. Although the majorityof LSCCs were positive for E-cadherin and only a smallpopulation were positive for Vimentin and SOX2, thesefactors showed considerable heterogeneity in TUM622-derived spheroids [62]. Because there were cells positivefor both E-cadherin and Vimentin, it is likely that partialEMT occurs in spheroids, the PDX model and the ori-ginal tumor [62, 65].Single cancer cell migration, also known as mesenchy-

mal migration, is characterized by fibroblast-like morph-ology, but effective metastasis of cancer cells can occur

without complete loss of epithelial morphology orcomplete acquisition of mesenchymal morphology. Can-cer cells undergoing mesenchymal migration areenriched at the invasive front in vivo, consistent withprevious findings that partial EMT is involved in collect-ive tumor migration [65, 66]. Leader cells expressingmesenchymal-like or basal epithelial traits are located atthe front of the follower epithelial cancer clusters, anddrive their collective migration in response to microenvi-ronmental cues. SOX2 appears to induce the commit-ment and differentiation of TUM622 cells to thesquamous lineage instead of regulating epithelial/mesen-chymal plasticity [62, 67]. SOX2 preferentially interactswith the transcription factor p63, as opposed to the tran-scription factor OCT4 in LSCCs, which is the preferredSOX2-binding partner in embryonic stem cells [68].FGFR1 accelerates tumor development without forcingcells toward a particular tumor subtype. By contrast,SOX2 appears to be critical in driving cells toward anaggressive and penetrant LSCCs phenotype [67, 69]. Fur-thermore, CAF-derived CD81-positive exosomesmobilize Wnt11 produced by breast carcinoma cells,thereby activating β-catenin-independent Wnt planarcell polarity (PCP) signaling, which promotes lung me-tastasis [70]. Chen et al. suggested that the β-catenin-dependent canonical Wnt signaling pathway and cancerstem cell marker SOX2 synergistically induce acinarmorphogenesis of TUM622 cells in three-dimensionalculture [62]. Further investigations are warranted to de-termine how SOX2 overexpression and co-culture withCAFs affects the β-catenin-independent PCP pathwaywhen spheroid cells derived from this PDX model oflung cancer acquire an invasive phenotype via partialEMT.

Malignant melanomaBoth intrinsic and acquired therapy resistance remains aserious challenge for the management of BRAF(V600E)-mutant malignant melanoma. Many resistant cases ex-hibit reactivation of MAPK and PI3K signaling in thepresence of MAPK-targeting agents. De novo lipogenesisis emerging as a central player in multiple oncogenicprocesses. Constitutive activation of the lipogenic path-way in tumor tissue is required for the synthesis of phos-pholipids, which function as essential building blocks ofmembranes and promote cell growth and proliferation.In vitro, a marked decrease in de novo lipogenesis wasobserved in all BRAF(V600E)-mutant therapy-sensitive,but not therapy-resistant, cell lines in the presence ofthe BRAF inhibitor vemurafenib [71]. Remarkably, BRAFinhibition induced only a moderate decrease in expres-sion of sterol regulatory element-binding protein-1(SREBP-1), a master regulator of lipid metabolism, anddid not significantly affect lipogenesis in therapy-

Yoshida Journal of Hematology & Oncology (2020) 13:4 Page 6 of 16

Page 7: Applications of patient-derived tumor xenograft models and tumor … · 2020-01-07 · REVIEW Open Access Applications of patient-derived tumor xenograft models and tumor organoids

resistant melanoma cells. To assess the therapeutic po-tential of these findings, Talebi et al. investigated the im-pact of SREBP-1 inhibition in an in vivo pre-clinicalBRAF(V600E)-mutant melanoma model. Talebi et al. se-lected a PDX that responded poorly to BRAF inhibitorsalone [71, 72]. SREBP-1 contributes to the anti-tumorresponse induced by BRAF inhibition, and SREBP-1 in-hibition sensitizes therapy-resistant melanoma cells toMAPK-targeting therapy. In vivo analysis of oxidativestress revealed that while fatostatin or vemurafenib treat-ment alone did not significantly increase lipid peroxida-tion, combined vemurafenib/fatostatin treatment greatlyenhanced lipid peroxidation [71]. Notably, SREBP-1 in-hibition enhances the sensitivity to BRAF inhibitors in apre-clinical PDX model of melanoma [71].

Ovarian cancerChoi et al. established PDX models derived from serousadenocarcinoma and chemoresistant carcinosarcoma toinvestigate the therapeutic potential of itraconazole, anorally bioavailable anti-fungal drug that inhibits the en-zyme lanosterol 14α-demethylase [73]. Previous reportsidentified itraconazole as a potent antagonist of theHedgehog (Hh) signal pathway, which is different fromthe pathway involved in the inhibitory effect of this drugon fungal sterol biosynthesis [74]. Systemically adminis-tered itraconazole suppresses Hh pathway activity andmedulloblastoma growth in a mouse allograft modelsimilar to other Hh pathway antagonists, and surpris-ingly, itraconazole inhibits the Hh pathway at serumlevels comparable with those of patients undergoinganti-fungal therapy [74, 75]. This is a typical example ofdrug repurposing, in which the anti-cancer properties ofmedications otherwise administered for non-malignantdisorders serve to develop new treatments strategies forcancer [58]. Mechanistically, itraconazole antagonizesthe Hh pathway component Smoothened (Smo) througha mechanism distinct from that of cyclopamine andother known Smo antagonists, and prevents ciliary accu-mulation of Smo mediated by Hh stimulation. Both theHh and mammalian target of rapamycin (mTOR) signal-ing pathways are associated with angiogenesis in thetumor microenvironment. In ovarian cancer PDXmodels, addition of itraconazole to paclitaxel signifi-cantly enhanced therapeutic efficacy compared withpaclitaxel monotherapy. Expression of CD31 andVEGFR2 (angiogenesis markers), Gli1 (hedgehog signal-ing downstream molecule), and S6K1 (mTOR pathway)were all decreased in tumors treated with paclitaxel anditraconazole combination therapy [73]. Itraconazole hassynergistic effects when used in combination withpaclitaxel-based chemotherapy, which is one of the mosteffective available chemotherapeutics for ovarian cancer.

Table 2 provides the summary of the latest importantpapers using PDX models and organoids.

Future challenges of PDX modelsWhile the incorporation of PDX models in cancer re-search brings some exciting improvements, PDXs haveimportant limitations that must be addressed to improvethe availability of PDX models for translational researchand pre-clinical investigations, including “co-clinical tri-als.” Issues that must be addressed or standardized to fa-cilitate wide use of PDX models include (a) the site forimplantation of original tumor fragments, (b) timecourse for PDX tumor tissue generation, (c) the engraft-ment rate, (d) replacement of human stroma with mur-ine stroma, (e) failure to evaluate the immune system,and (f) the challenging problems of Matrigel.

Optimal implantation siteIt is important to define the best engraftment site (sub-cutaneous, subrenal capsule, or orthotopic implantation)in each tumor type of interest. Most of the publishedstudies using PDX models have relied on surgical speci-mens, which naturally provide large quantities of tumortissues. Although much effort has been expended in es-tablishing PDX models of cholangiocarcinoma and headand neck squamous cell carcinoma, acquiring a sufficientvolume for transplantation is prohibitive due to thesmall size of the original tumor. Thus, suitable methodswith smaller samples obtained by biopsy or fine-needleaspirations should be established.

Time course of PDX tumor tissue generationDelay between the engraftment period in mice and opti-mal treatment schedules for patients is a limiting factorfor the use of PDX models in real-time personalizedmedicine applications. Developing a PDX model for pre-clinical study generally requires 4–8 months with severaltumor passages, which is much longer than patients canordinarily wait to commence treatment. The second-and third-generation PDX passages require only 10 daysto form a palpable xenograft. Due to the time require-ment to generate PDX models, some groups are usingshort-term single-cell suspension and short-term culturein organoid models to evaluate sensitivity to potentialtreatments.

Tumor engraftment rateThe engraftment failure rate is still high for some cancertypes and phenotypes, which is a major obstacle to wide-spread PDX use. It is thus essential to improve tumorengraftment rates to an acceptable level, up to 60–70%.Most importantly, patients with breast and renal cancerwhose tumors successfully engraft have the worst prog-nosis [4, 5, 103], which strongly suggests a selective

Yoshida Journal of Hematology & Oncology (2020) 13:4 Page 7 of 16

Page 8: Applications of patient-derived tumor xenograft models and tumor … · 2020-01-07 · REVIEW Open Access Applications of patient-derived tumor xenograft models and tumor organoids

Table

2Recent

investigations

ofsolid

tumorsusingPD

Xmod

els

Tumor

type

Reference

Animal

Site

Metastasis

Colon

aden

ocarcino

ma

Misaleet

al.[12]

NOD-SCID

mice

Subcutaneo

usLiver

PDXmod

elsde

rived

from

aqu

adruplewild-type(KRA

S,NRA

S,BRAF,and

PIK3CA

)colorectaltumor

aresensitive

toEG

FRblockade

alon

e.EG

FR/M

EKcombinatio

nblockade

issupe

riorto

treatm

entwith

anti-EG

FRantib

odies,or

tode

liveringaMEK

inhibitorwhe

nresistance

tocetuximab

orpanitumum

abhasalreadyde

velope

d.

Tumor

type

Reference

Animal

Site

Metastasis

Colon

aden

ocarcino

ma

Fujiiet

al.[24]

NOGmice

Subren

alcapsuleand

spleen

Liver

Fujiiet

al.establishe

dacolorectaltumor

organo

idlibrary

comprised

of55

organo

idsde

rived

from

52tumorsand43

patients,includ

inghype

rplasticpo

lyps

andsessile

serrated

aden

oma/po

lyps.Patho

histolog

ical

subtypes

aswellasdifferentiatio

nhierarchiesof

CRC

swerecell-intrinsically

conservedregardless

ofen

vironm

ent(i.e.,inpatients,in

vitro,

andin

PDXmod

els).PDXmod

elsestablishe

dby

transplantationinto

the

kidn

eysubcapsulesof

immun

ocom

prom

ised

micede

velope

dthesize

ofen

graftedsubren

alCRC

sthat

secretenichefactors,includ

ingp3

8-MAPK,TGF-β,

andEG

F.In

contrastto

therobu

sten

graftm

entefficiency

insubren

alcapsules,the

metastatic

capacity

ofspleen

-injected

CRC

organo

idswas

diverse.

Tumor

type

Reference

Animal

Site

Metastasis

Colon

aden

ocarcino

ma

Fior

etal.[17]

Zebrafish

Subcutaneo

usNon

e

Fior

etal.g

enerated

andtreatedfivezebrafish-basedPD

X(zPD

X)mod

elsof

coloncancer

derived

from

different

patients,andtreatedzPDXs

with

theFO

LFOXregimen

over

3days.TwoPD

Xsrespon

dedto

treat-

men

t,as

indicatedby

increasedcaspase3cleavage

.The

setw

osensitive

zPDXs

correspo

nded

topatientsin

who

mCEA

levelsremaine

dstable6mon

thsaftersurgerywith

outrelapse.Con

trastin

gly,am

ongthe

threezPDXs

inwhich

FOLFOXwas

noteffective,thecorrespo

ndingpatientsde

velope

dincreasing

CEA

levelsandclinicaleviden

ceof

relapse.Furthe

rmore,Fior

etal.investig

ated

thepred

ictiveeffect

oftheEG

FRinhibitorCetuxim

abin

combinatio

nwith

FOLFIRIreg

imen

,finding

that

resistantzPDXmod

elsarede

rived

from

tumorswith

mutations

ineither

BRAF

orKRAS.

Tumor

type

Reference

Animal

Site

Metastasis

Colon

aden

ocarcino

ma

Okazawaet

al.[56]

NOGmice

Ortho

topic

Lung

s

Pieces

ofresected

CRC

sweresubcutaneo

uslyim

plantedinto

NOGmiceto

gene

rate

thePD

Xmod

el.The

organo

idcells

werethen

extractedfro

mthePD

Xmod

elfortissuecultu

re,and

CRC

organo

idswere

infected

with

GFP

lentiviru

s,allowinghigh

lysensitive

visualizationof

micrometastases(Fig.2).Notably,lun

gmicrometastaseswerede

tected

inthreeou

tof

four

miceexam

ined

2.5mon

thsafterorthotop

icinjectionof

GFP-labe

lledPD

X-de

rived

organo

ids.Theim

plantatio

nof

organo

idsinto

therectalsubm

ucosaof

NOGmiceresultedin

metastatic

dissem

inationinto

thelung

s,bu

tno

ttheliver,p

resumablythroug

htheinferio

rvena

cava.

Tumor

type

Reference

Animal

Site

Metastasis

Pancreaticcancer

Zhou

etal.[76]

Nud

emice,SC

IDmice

Ortho

topic

Non

e

Becauseinsulin

grow

thfactor

1receptor

(IGF1R)

ishigh

lyexpressedin

both

pancreaticcancer

cells

andstromalfib

roblasts,Z

houet

al.d

evelop

ednano

particleswith

recombinant

human

IGF1

conjug

ated

tomagne

ticiro

noxidecarrying

anthracyclinedo

xorubicin(IG

F1-IO

NP-Dox),andde

mon

stratedan

enhanced

therapeutic

effect

comparedwith

conven

tionalD

oxtreatm

entin

theorthotop

icpancreaticdu

ctal

aden

ocarcino

ma(PDAC)PD

Xmod

el.T2-weigh

tedmagne

ticresonanceim

aging(M

RI)revealed

system

icde

liveryof

IGF1R-targeted

Dox

afteradministrationof

IGF1-IO

NP-Dox.N

otably,n

on-spe

cific

uptake

ofIGF1-

IONP-Dox

inthespleen

didno

tcauseapop

tosis,as

demon

stratedby

lack

ofactivecaspase3.

Tumor

type

Reference

Animal

Site

Metastasis

Pancreaticcancer

Witkiewiczet

al.[77]

NSG

mice

Subcutaneo

usNon

e

PDXmod

elsof

PDACen

ableprecisiontherapy,as

resistance

toMEK

inhibitorsisparado

xically

associated

with

compe

nsatoryAkt

sign

alingactivation[78].W

itkiewiczet

al.usedthismod

elto

demon

strate

that

combinatio

ntherapywith

tram

etinib

(MEK

inhibitor)anddasatin

ib(tyrosinekinase

Srcinhibitor)sign

ificantlysupp

resses

PDXtumor

proliferatio

n.Furthe

rmore,theBcl-2

inhibitorABT737andcheckpoint

kinase

in-

hibitorAZD

7762

indu

cesatherapeutic

respon

sein

coop

erationwith

conven

tionalanti-cancerdrug

ssuch

asdo

cetaxeland

gemcitabine

.

Tumor

type

Reference

Animal

Site

Metastasis

Pancreaticcancer

Rajeshkumar

etal.[79]

Nud

emice

Subcutaneo

usNon

e

PDXmod

elsof

PDACrespon

dmorerobu

stlyto

mito

chon

drialcom

plex

Iinh

ibito

rs(phe

nformin

andmetform

in)than

toothe

rmetabolicinhibitors,including

aglutam

inaseinhibitor,atransaminaseinhibitor,and

anautoph

agyinhibitor.Aminoacidsandmetabolitesinvolved

inglycolysissuch

aslactatearede

creasedby

complex

Iinh

ibito

rs,w

hileoxidized

glutathion

eisincreased.

Thereisno

correlationbe

tween

phen

form

inrespon

seandge

netic

abno

rmalities

inTP53,PTEN,SMAD

4,andKRAS.A

lthou

ghph

enform

inhasno

tbe

enappliedclinicallydu

eto

theriskof

lacticacidosis[80],thisstud

ysugg

eststheclinicalprom

ise

ofthisbigu

anideagen

t.

Tumor

type

Reference

Animal

Site

Metastasis

Yoshida Journal of Hematology & Oncology (2020) 13:4 Page 8 of 16

Page 9: Applications of patient-derived tumor xenograft models and tumor … · 2020-01-07 · REVIEW Open Access Applications of patient-derived tumor xenograft models and tumor organoids

Table

2Recent

investigations

ofsolid

tumorsusingPD

Xmod

els(Con

tinued)

Cho

lang

iocarcinom

aGarciaet

al.[14]

SCID

mice

Subcutaneo

usNon

e

Five

PDXmod

elsof

cholangiocarcino

maexhibitediden

ticalKRAS

mutations

tothoseof

thetumorsfro

mwhich

they

werede

rived

.Ind

eed,

theon

coge

nicKRAS

mutationfre

quen

tlyoccursas

apo

intmutationin

codo

n12,and

thesemutations

resultin

constitutiveactivationof

thePI3K

pathway

andRA

S/RA

F/MEK/ERK

axis,w

hich

prom

otecancer

cellsurvivalandproliferatio

n[81].C

ell-cycleregu

latory

proteins,including

Chk1andE2F1,w

ereselectivelydo

wn-regu

latedby

BETproteininhibitorJQ

1in

JQ1-sensitive

PDXmod

els.WhileJQ

1failedto

affect

c-Myc

expression

inJQ

1-insensitive

PDXmod

els,aBETinhibitordo

wn-

regu

latedc-Myc

inJQ

1-sensitive

PDXtumors,which

was

accompanied

bydo

wn-regu

latio

nof

downstream

transcrip

tionaltarge

tssuch

asChk1andE2F1.

Tumor

type

Reference

Animal

Site

Metastasis

Glioblastomamultiforme

Leeet

al.[82]

Nud

emice

Ortho

topic

Non

e

Leeat

al.d

evelop

edGBM

recurren

tPD

Xmod

elsindu

cedby

temozolom

ide,in

which

theim

med

iate

peak

stabilizatio

nof

HIF1α

inPD

Xcells

afterexpo

sure

tochem

othe

rapy

isexpe

cted

torepresen

tan

essential

step

intheconversion

ofno

n-cancer

stem

cells

into

undifferentiatedcancer

stem

cells

(CSC

s).Inbo

thGBM

6(classical,M

GMThype

rmethylated),and

GBM

43(prone

ural,M

GMTun

methylated)

PDXmod

els,HIF1α

levelswereincreasedin

theCD133-po

sitiveCSC

popu

latio

nbo

thpo

st-the

rapy

andat

diseaserecurren

ce.

Tumor

type

Reference

Animal

Site

Metastasis

Glioblastomamultiforme

Sing

het

al.[59]

NOD-SCID

mice

Ortho

topic

Non

e

Ectopicco-expressionof

SOX2

,OLIG2,andZEB1

transformstumor-sup

pressor-de

ficient

astrocytes

into

glioma-initiatingcells

intheabsenceof

anup

stream

RTKon

coge

ne.A

mon

gthethreetranscrip

tionfactors,

SOX2

expression

issign

ificantlyup

regu

latedin

PDXGBM

s.Histone

H3lysine

27residu

e(H3K27)acetylationwas

presen

tin

morethan

90%

ofthePD

XSO

X2bind

ingregion

sin

thethreeanalyzed

patient

GBM

specim

ens,which

indicatestheseregion

sas

activecis-regu

latory

elem

entsin

patient

GBM

.

Tumor

type

Reference

Animal

Site

Metastasis

Glioma

Fack

etal.[83]

NOD-SCID

mice

Ortho

topic

Non

e

Fack

etal.app

liedin

situ

metabolicprofiling

andLC

-MSon

brainsections

ofgliomaPD

Xandhu

man

gliomasamples

with

andwith

outisocitratede

hydrog

enase1(ID

H1)

mutations.M

assspectrom

etry

imaging

(MSI)andLC

-MSanalysisof

orthotop

icIDH-m

utated

gliomaPD

Xmod

elsrevealed

IDH-spe

cific

adaptivemechanism

sin

metabolicpathways.Notably,cystathionine

-β-synthaseexpression

isano

velp

rogn

ostic

fac-

torin

theoligod

endrog

lialg

liomasubtype.

Tumor

type

Reference

Animal

Site

Metastasis

Med

ulloblastoma

Garne

ret

al.[84]

Nud

emice

Ortho

topic

Non

e

Threegrou

p3med

ulloblastomaPD

Xmod

elswereestablishe

dto

evaluate

thetherapeutic

efficacyof

FTY720,anim

mun

osup

pressant

that

hascurren

tlybe

enapproved

fortreatm

entof

multip

lesclerosis[85].

FTY720

activates

thetumor

supp

ressor

proteinph

osph

atase2A

,and

thus

treatm

entof

human

med

ulloblastomaPD

Xcells

with

FTY720

arrested

thecellcycleat

theG1ph

aseandindu

cedcaspase-de

pend

ent

apop

totic

cellde

ath.

Tumor

type

Reference

Animal

Site

Metastasis

Breastcarcinom

aLawsonet

al.[8]

NOD-SCID

mice

Ortho

topic

Lung

s,bo

nemarrow,liver,b

rain

Inthisstud

y,themostmetastatic

PDXmod

el(HCI-010)exhibitedthehigh

estpe

rcen

tage

ofbasal/stem-like

tumor

cells,w

hiletheleastmetastatic

mod

el(HCI-002)hadthelowest.Thissugg

eststhat

prim

ary

tumorscontainarare

subp

opulationof

stem

-like

cells,and

that

therelativeabun

danceof

thesecells

correlates

with

metastatic

potential.Highe

r-bu

rden

metastatic

cells

enteredthecellcycle,expressing

lower

levelsof

quiescen

ceanddo

rmancy-associatedge

nesandhigh

erlevelsof

cell-cycle-prom

otingge

nes,includ

ingCD24,C

DK2,M

MP1,and

MYC

,which

have

been

associated

with

reactivationafterdo

rmancy.D

inaci-

clib,a

CDKinhibitorthat

indu

cesapop

tosisin

high

MYC

-expressingcancer

cells

viasynthe

ticlethality

[86,87],sign

ificantlyinhibitsthemetastatic

potentialo

fPD

Xmod

elsde

rived

from

drug

-naïve

patients.

Tumor

type

Reference

Animal

Site

Metastasis

Breastcarcinom

aEvanset

al.[13]

Nud

emice

Ortho

topic

Non

e

Morethan

25PD

Xmod

elsde

rived

from

triple-neg

ativebreastcancer

(TNBC

)tissues

variedin

theextent

ofPI3K

andMAPK

activation.PD

Xswerealso

heteroge

neou

sin

theirsensitivity

tochem

othe

rape

uticagen

ts;

whilePI3K,m

TOR,andMEK

inhibitorsrepressedgrow

thbu

tdidno

tcausetumor

regression

,the

PARP

inhibitortalazoparib

caused

drastic

regression

infiveof

12PD

Xs.N

otably,fou

rou

tof

fivetalazoparib

-sensitive

mod

elsdidno

tharbor

germ

lineBRCA

1/2mutations,b

utseveralh

adsomaticalteratio

nsin

homolog

ousrepairpathways,includ

ingATM

deletio

nandBRCA

2alteratio

ns.

Tumor

type

Reference

Animal

Site

Metastasis

Breastcarcinom

aYu

etal.[88]

NOD-SCID

mice

Subcutaneo

usNon

e

Toelucidatethemechanism

of5-Aza-2'-d

eoxycytid

ine(decitabine

)actio

non

TNBC

,Yuet

al.investig

ated

DNAmethyltransferases

(DNMTs)expression

levels,w

hich

werecorrelated

with

respon

seto

decitabine

inbreastcancer

organo

idmod

elsbasedon

PDXtumorsde

rived

from

TNBC

patientsrecruitedto

aprospe

ctivene

oadjuvantstud

yof

anthracycline-

andtaxane

-based

chem

othe

rapy.O

rganoids

andPD

Xmod

elsex-

pressing

elevated

levelsof

DNMTproteins

weremoresensitive

tode

citabine

than

thoseexpressing

low

levelsof

DNMTs,sug

gestingthat

DNMTmay

beapred

ictivebiom

arkerfortreatm

entrespon

seto

decitabine

inTN

BC.Thiseffect

was

med

iatedby

decitabine

-indu

cedub

iquitin

ationandlysosomalde

gradationof

allthree

DNMTs,w

hich

was

depe

nden

ton

theE3

ligaseTN

Freceptor-associatedfactor

6(TRA

F6).Decitabine

Yoshida Journal of Hematology & Oncology (2020) 13:4 Page 9 of 16

Page 10: Applications of patient-derived tumor xenograft models and tumor … · 2020-01-07 · REVIEW Open Access Applications of patient-derived tumor xenograft models and tumor organoids

Table

2Recent

investigations

ofsolid

tumorsusingPD

Xmod

els(Con

tinued)

treatm

entof

PDXtissues

also

increasedTRAF6transcrip

tion.

Tumor

type

Reference

Animal

Site

Metastasis

Breastcarcinom

aXiao

etal.[9]

NSG

mice

Subcutaneo

usNon

e

p21protein–

activated

kinase

2(PAK2)isactivated

bylow

C-terminalSRCkinase

levels,w

hich

drives

estrog

en-in

depe

nden

ttumor

grow

thin

patientswith

estrog

enreceptor

(ER)–p

ositive

breastcancer

resistantto

endo

crinetherapy.Using

aPD

Xmod

elde

rived

from

ER(+)/Pg

R(+)/HER2(-)invasive

ductalcarcinom

a,Xiao

etal.con

firmed

that

combinatio

ntreatm

entwith

thePA

K2inhibitorFRAX5

97andan

ERantago

nistsyne

r-gisticallysupp

ressed

breasttumor

grow

th.

Tumor

type

Reference

Animal

Site

Metastasis

Breastcarcinom

aIkbaleet

al.[89]

NSG

mice

Ortho

topic

Lymph

node

s,visceralmetastasis

Com

paredwith

treatm

ent-naïveTN

BC–d

erived

PDX,

chem

oresistant

TNBC

–derived

PDXhigh

lyexpressedWnt10B-relatedmolecules,including

non-ph

osph

orylated

activeβ-catenin,

Axin2

,CD44,and

HMGA2.ICG-

001,acano

nicalW

ntsign

alinginhibitor,redu

cedtumor

grow

thandlymph

node

metastatic

burden

.The

combinatio

nof

doxorubicinandICG-001

efficientlyrepressedlung

dissem

inationaftertailvein

injectionof

tumor

cells

that

haddissociatedfro

mchem

oresistant

TNBC

PDX.

Thissyne

rgistic

effect

was

med

iatedby

Bcl-2

downreg

ulation.

Tumor

type

Reference

Animal

Site

Metastasis

Lung

cancer

Weede

net

al.[90]

NSG

mice

Subcutaneo

usNon

e

PDXmod

elsde

rived

from

lung

squamou

scellcarcinom

aen

abledWeede

net

al.toiden

tifyFG

FR1mRN

Alevelrathe

rthan

FGFR1am

plificatio

nas

aprecisepred

ictorof

respon

seto

anFG

FRtyrosine

kinase

inhibitor.Seventeenpe

rcen

tof

PDXs

evaluated(sixou

tof

36cases)exhibitedFG

FR1am

plificatio

n,andFG

FRinhibitio

nde

creasedcellproliferatio

nanden

hanced

differentiatio

n,which

decreasedtumor

grow

thrate

andon

lymod

estly

increasedapop

totic

cellde

ath.

Treatm

entwith

theFG

FRinhibitorBG

J398

blockedbo

thAKT

andERKsign

aling,

andcombinatio

ntherapywith

BGJ398

andaPI3K

inhibitorwas

bene

ficial

onlyin

PDXs

inwhich

FGFR

inhibitio

ndidno

talterPI3K

sign

aling.

Tumor

type

Reference

Animal

Site

Metastasis

Lung

cancer

Drapkin

etal.[91]

NSG

mice

Subcutaneo

usNon

e

Becausecollectionof

circulatingtumor

cells

(CTC

s)en

ablesno

n-invasive

serialtum

orsampling,

Drapkin

etal.establishe

dmorethan

30PD

Xmod

elsof

smallcelllun

gcancer

(SCLC

)de

rived

from

noton

lybiop

sytissues

butalso

from

CTC

s.SC

LCPD

Xmod

elsretain

stablege

nomesomaticalteratio

nsbe

tweeninitialPD

Xmod

elge

neratio

nandserialp

assage

s.Surprisingly,CTC

-derived

PDXmod

elsreflect

theevolving

chem

o-therapysensitivitiesof

theoriginaltumor

atthetim

eof

CTC

collection.

Etop

osideresistance

ispo

sitivelycorrelated

with

activationof

theMyc-associatedge

nesign

ature,which

isconsistent

with

results

obtained

usingthege

neticallyen

gine

ered

mou

semod

elof

SCLC

[92,93].In

comparison

toothe

rmalignancies,SC

LCPD

Xmod

elsexhibitincreasedclon

alho

mog

eneity

andge

nomicstability.

Tumor

type

Reference

Animal

Site

Metastasis

Lung

cancer

Gon

get

al.[94]

NOD-SCID

mice

Subcutaneo

usNon

e

Inge

neral,patientswith

wild

type

EGFR

(EGFRwt)do

notrespon

dto

treatm

entwith

EGFR

tyrosine

kinase

inhibitors(TKIs),w

hilemostpatientswith

EGFR

activatingmutations

initiallyrespon

dto

EGFR

TKIs,b

utinevitablyde

velopsecond

aryresistance

toTKItreatmen

t.Simultane

ousinhibitio

nof

EGFR

andTN

Fpreven

tsde

velopm

entof

thisacqu

iredEG

FRresistance.N

otably,com

binatio

ntreatm

entwith

EGFR

TKI

(erlo

tinib)plus

thalidom

idewas

high

lyeffectivein

inhibitin

gtumor

grow

thin

anEG

FRwtPD

Xmod

el,w

hileEG

FRinhibitio

nor

thalidom

idealon

ewas

ineffective.

Tumor

type

Reference

Animal

Site

Metastasis

Lung

cancer

Che

net

al.[62]

Nud

emice

Subcutaneo

usNon

e

TheTU

M622celllineestablishe

dfro

mPD

Xsof

lung

squamou

scellcarcinom

ahasincreasedsphe

roid-fo

rmingcapacity

dueto

overexpression

ofthestem

cellfactor

SOX2

,and

show

sahype

rplasticto

dysplastic

change

inits

acinar

phen

otype,in

which

apical-basalcellpo

larityisdisrup

ted.

Wnt/β-caten

insign

alingandSO

X2,w

hich

contrib

uteto

norm

allung

developm

ent,areinvolved

inacinar

morph

ogen

esisof

TUM622

cells

inthree-dimen

sion

alcultu

res.Che

net

al.rep

orteden

hanced

epith

elial/m

esen

chym

alplasticity

ofTU

M622cells

afterincorporatingcancer-associatedfib

roblasts(CAFs)into

asphe

roid

cultu

resystem

,and

expand

ingthissystem

tomod

elno

ton

lycancer

cellEC

M,b

utalso

cancer

cell-CAFinteractions

durin

gtumorigen

esis.C

AFs

antago

nizedon

coge

nicSO

X2to

restoretheform

ationof

luminalstructures

andpro-

moteinvasion

.

Tumor

type

Reference

Animal

Site

Metastasis

Malignant

melanom

aHirata

etal.[95]

NSG

mice

Subcutaneo

usLung

s

Intravitalimagingof

BRAF-m

utantmelanom

acells

containing

anERK/MAPK

biosen

sorrevealed

how

thetumor

microen

vironm

entaffectsrespon

sesto

BRAFinhibitio

nby

PLX4

720.Inde

ed,BRA

F-mutantmelanom

acells

respon

dto

PLX4

720he

teroge

neou

sly,andBRAFinhibitio

nactivates

CAFs,leading

toFA

K-de

pend

entmelanom

asurvivalsign

aling.

Fibron

ectin

-richmatrices

with

3-12

kPaelastic

mod

ulus

aresufficien

tto

in-

duce

PLX4

720tolerance.ThePD

Xmod

elrevealed

that

co-in

hibitio

nof

BRAFandFA

Kabolishe

sERKreactivationin

tumor

stroma,leadingto

moreeffectivecontrolo

fBRAF-m

utantmelanom

a.

Tumor

type

Reference

Animal

Site

Metastasis

Yoshida Journal of Hematology & Oncology (2020) 13:4 Page 10 of 16

Page 11: Applications of patient-derived tumor xenograft models and tumor … · 2020-01-07 · REVIEW Open Access Applications of patient-derived tumor xenograft models and tumor organoids

Table

2Recent

investigations

ofsolid

tumorsusingPD

Xmod

els(Con

tinued)

Malignant

melanom

aKrep

leret

al.[96]

NSG

mice

Subcutaneo

usBrain,

lung

s

Tofacilitatetheadvancem

entof

pre-clinicalin

vivo

mod

eling,

Krep

leret

al.establishe

dmorethan

450PD

Xmod

els.Half(55%)of

allanalyzedsamples

wereBRAF

hotspo

tmutants,20%

wereNRA

Smutants,7%

wereNF1

mutants,2%

wereKITmutants,1.4%

wereGNAQ

/GNA11mutants,and

18%

wereWT,allo

fwhich

wereconsistent

with

theTC

GAdatabase.The

simultane

ousinhibitio

nof

MDM2andERKishigh

lyeffect-

ivein

PDXs

refractoryto

BRAFinhibitors.

Tumor

type

Reference

Animal

Site

Metastasis

Malignant

melanom

aTalebi

etal.[71]

Nud

emice

Subcutaneo

usNon

e

SREBP-1protectsvemurafen

ib-resistant

melanom

acells

from

lipid

peroxidatio

n.Fatostatin

treatm

entalon

eof

PDXMEL006inhibitstumor

grow

thmorepo

tentlythan

vemurafen

ib.Impo

rtantly,com

bine

dvemura-

fenib/fatostatin

co-treatmen

thasagreateranti-tumor

effect

than

either

mon

othe

rapy

regimen

.

Tumor

type

Reference

Animal

Site

Metastasis

Malignant

melanom

aEinarsdo

ttiret

al.[97]

NOGmice

Subcutaneo

usNon

e

PDXmod

elsde

rived

from

metastatic

melanom

awereestablishe

dto

assess

heteroge

neou

srespon

sesaftertreatin

gtumorswith

karonu

dib,

which

inhibitstheoxidized

nucleo

tide-sanitizingen

zymeMTH

1[98].

Com

parison

ofthemutationprofile

betw

eenrespon

segrou

psrevealed

that

karonu

dibhasacytotoxiceffect

inmelanom

aPD

Xmod

els,irrespe

ctiveof

themutationstatuses

ofthemostcommon

driver

gene

sin

melanom

a.Im

portantly,h

ighexpression

ofABC

B1isapo

tentialresistancebiom

arker.

Tumor

type

Reference

Animal

Site

Metastasis

Headandne

cksquamou

scellcarcinom

a(HNSC

C)

Grasset

etal.[99]

NMRI-nud

emice

Subcutaneo

usNon

e

ThePD

Xmod

elrevealed

that

theLO

XinhibitorBA

PNattenu

ated

ECM

remod

elingandtumor

stiffne

sswith

collage

nIb

undles

byredu

cing

EGFR

tyrosine

kinase

activity

ofcancer

cells.The

Ca2

+channe

lblockers

phen

ylalkylamineverapamilandno

ndihydropyrid

inediltiazem

,usedforthetreatm

entof

hype

rten

sion

andarrhythm

iaforde

cade

s,areeffectivein

PDXmod

elsforthepreven

tionof

collectivetumor

cellinvasion

bysign

ificantlydo

wnreg

ulatingph

osph

orylated

myosinlight

chain2.Mechanistically,tum

or-derived

ECM

stiffne

ssandactivated

EGFR

sign

alingen

hances

intracellularCa2

+concen

trationmed

iatedby

theL-type

Ca2

+channe

lCav1.1in

squamou

scancer

cells.

Tumor

type

Reference

Animal

Site

Metastasis

Ovariancancer

Liuet

al.[100]

Nud

emice,NSG

mice

Intraperito

nealspace

Periton

eald

isseminationsuch

asom

entum,liver,p

ancreas,bo

wel,spleenand

diaphragm

Liuet

al.establishe

dPD

Xmod

elsof

ovariancarcinom

ade

rived

from

floatingtumor

cells

intheascitesof

irradiatednu

demice,andascitesin

establishe

dPD

Xmod

elswerethen

implantedintraperito

neallyinto

NSG

mice.Freshascites-de

rived

tumor

cells

from

thesePD

Xtumor-bearin

gNSG

miceweretransfectedwith

lentiviru

sen

coding

fireflyluciferase

andmChe

rryforbiolum

inescenceim

aging(BLI)of

PDXmod

els.Co-

hortsof

NSG

micewith

luciferized

PDXtumorsweretreatedwith

carbop

latin

,orpaclitaxel,either

asamon

othe

rapy

orin

combinatio

n,followed

byweeklyBLIm

easuremen

ts,the

results

ofwhich

positivelycorre-

latedwith

thoseof

plasmaCA125andcell-fre

eDNAassays.N

otably,PDXmod

elsde

rived

from

platinum

-refractorypatientsde

mon

stratedsign

ificant

resistance

tocarbop

latin

.

Tumor

type

Reference

Animal

Site

Metastasis

Ovariancancer

Kim

etal.[101]

NSG

mice

Ortho

topic

Non

e

Kim

etal.investig

ated

thetherapeutic

effectsof

aPA

RPinhibitor(PARPi;olaparib),an

ATR

inhibitor(ATRi;AZD

6738),andaCHK1

inhibitor(CHK1i;MK8776)

forthetreatm

entof

BRCA

-mutanthigh

-grade

serous

ovariancancer

(HGSO

C)cells.A

lthou

ghPA

RPiand

CHKias

sing

leagen

tsmod

estly

supp

ressed

tumor

grow

th,the

additio

nof

ATRi/C

HK1itoPA

RPiinBRCA

-mutantovarianPD

Xmod

elssign

ificantlyde

creased

tumor

volumecomparedwith

sing

le-age

nttherapies.Remarkably,sign

ificant

differences

wereob

served

inrespon

sivene

ssto

thePA

RPi-A

TRiand

PARP

i-CHK1icom

binatio

ns.W

hilePA

RPi-C

HK1icom

binatio

ntreat-

men

tsign

ificantlyincreasedPD

Xtumor

supp

ressionover

sing

le-age

nttreatm

ents,PARPi-A

TRicom

binatio

ntreatm

entsign

ificantlyincreasedtheincide

nceof

PDXtumor

regression

.

Tumor

type

Reference

Animal

Site

Metastasis

Ovariancancer

Geo

rgeet

al.[18]

NSG

mice

Ortho

topic

Diaph

ragm

PDXmod

elsde

rived

from

BRCA

-mutantHGSO

Cspatholog

icallyexhibitedsolid,p

seud

oend

ometrio

id,and

transitio

nalcellcarcino

mas,referredto

asSETfeatures.Because

approxim

ately50%

ofHGSO

Cpatients

have

defectsin

homolog

ousrecombinatio

n(i.e.,loss-of-fu

nctio

nalm

utations

ofBRCA

1/2)

[102],blocking

cellcyclecheckpointscouldpo

tentially

indu

cesynthe

ticlethality

inHGSO

C.Ind

eed,

PDXmod

elsof

BRCA

2-mutantHGSO

Cexhibitedhigh

erlevelsof

phosph

orylated

CHK1

than

BRCA

-intact

HGSO

C.PET

imagingof

PARP-1

with

thePA

RPinhibitoranalog

ue[18 F]FTTwas

perfo

rmed

inPD

Xmod

els,which

revealed

that

PARP

inhibitortreatm

entresultedin

tumor

supp

ressionbu

tno

tcompletetumor

regression

,sim

ilarto

therespon

seob

served

inclinicalsettings.

Tumor

type

Reference

Animal

Site

Metastasis

Ovariancancer

Cho

ietal.[73]

Nud

emice

Subren

alcapsule

Non

e

Cho

ietal.examined

thetherapeutic

effect

oftheantifun

galitracon

azolein

PDXmod

elsof

serous

aden

ocarcino

maandcarcinosarcoma.Com

binatio

ntherapywith

paclitaxeland

itracon

azolesign

ificantly

Yoshida Journal of Hematology & Oncology (2020) 13:4 Page 11 of 16

Page 12: Applications of patient-derived tumor xenograft models and tumor … · 2020-01-07 · REVIEW Open Access Applications of patient-derived tumor xenograft models and tumor organoids

Table

2Recent

investigations

ofsolid

tumorsusingPD

Xmod

els(Con

tinued)

inhibitedtumor

grow

thandsupp

ressed

angiog

enesis.C

ombinatio

ntherapywas

moreeffectivein

decreasing

CD31,VEG

Freceptor,G

li1-m

ediatedHh,

andmTO

Rsign

alingthan

paclitaxelm

onothe

rapy.

Tumor

type

Reference

Animal

Site

Metastasis

Ovariancancer

Kond

rashovaet

al.(2018)

[49]

NOGmice

Subcutaneo

usand

orthotop

icNon

e

Therespon

seof

PDXmod

elsto

thePA

RPinhibitorrucaparib

largelyde

pend

son

BRCA

1prom

oter

methylatio

nzygo

sity.PDXmod

elswerede

rived

from

12HGSO

Cpatients,tenof

who

mwerechem

othe

rapy-

naïve,andtw

oof

who

mhadreceived

multip

lelines

oftherapy.Ahe

terozygo

ustherapeutic

respon

seto

rucaparib

was

observed

amon

gBRCA

1/2-mutantHGSO

CPD

Xmod

els.Ko

ndrashovaet

al.o

bservedaltered

zygo

sity

ofBRCA

1methylatio

n,characterized

byho

mozygosity

inthechem

o-naïvepatient-derived

samples

andhe

terozygo

sity

inthepreviouslytreatedHGSO

Cpatient–d

erived

samples.Lossof

homozygou

sBRCA

1methylatio

nislikelyrespon

siblein

partforacqu

iredrucaparib

resistance.

Tumor

type

Reference

Animal

Site

Metastasis

Bladde

rcancer

Leeet

al.(2018)[50]

NOGmice

Ortho

topic

Muscleinvasion

Leeet

al.d

evelop

edan

optim

ized

metho

dology

toconvertbladde

rtumor

organo

idlines

into

orthotop

icPD

Xmod

elswith

upto

80%

efficiencyusingultrasou

nd-guide

dim

plantatio

nof

organo

idsbe

tweenthe

bladde

rurothe

lium

andlaminaprop

ria.Tum

orevolutioniscommon

inorgano

ids,even

intheabsenceof

drug

treatm

ent.Patient-derived

bladde

rcancer

organo

idsmaintaine

dhe

teroge

neity,leading

toclon

alevolution.

Notably,the

basalo

rganoidph

enotypereversiblychange

dinto

theluminalph

enotypein

PDXmod

els,likelydu

eto

cellularplasticity.PDXmod

elsretain

drug

respon

sesto

tram

etinib

(MEK

inhibitor)

andge

mcitabine

.

Yoshida Journal of Hematology & Oncology (2020) 13:4 Page 12 of 16

Page 13: Applications of patient-derived tumor xenograft models and tumor … · 2020-01-07 · REVIEW Open Access Applications of patient-derived tumor xenograft models and tumor organoids

pressure towards more aggressive phenotypes. It is pos-sible that as the passages of the PDX model proceed,proliferative, and highly metastatic clones are selected toestablish the next generation of PDXs.

Replacement of human stroma with murine stromaIntratumoral heterogeneity can be influenced by tumor-extrinsic factors in the microenvironment, includingmurine host cells. After 3–5 passages, when PDX modelscan be used for drug screening, tumor-associated stromaare almost completely replaced by murine-derived ECMand fibroblasts. This new murine stroma is likely tocause drastic changes in paracrine regulation of thetumor as well as in physical properties such as intersti-tial fluid pressure, which disturb drug distribution [104].Importantly, some cytokines are species-specific. Asstated previously, human-derived IL-2 stimulates theproliferation of murine T cells at similar concentrations,whereas mouse IL-2 stimulates human T cells at a sig-nificantly lower efficacy [45]. Furthermore, the T cellstimulating potential of IL-4 seems to be species specific.IL-15 binds to human or mouse IL-15 receptor α withequal high affinity [47]. Surely human-derived IL-15 canfunction on murine cells; human NK cells are poorlysensitive to murine IL-15 [48].

Failure to evaluate the immune systemOne key requirement in establishing PDX models is theneed to use immunodeficient host strains for tumor en-graftment and propagation. These mice lack functionalelements of the immune system, so that foreign tumortissues cannot be rejected. For example, NSG mice lacknatural killer cells and both B and T lymphoid cells.Therefore, the contribution of the host immune systemto responses to conventional chemotherapeutics cannotbe assessed. Currently available PDX models are also un-able to accurately assess immunotherapies such as vac-cines and immune modulators, and drugs that activatethe anti-tumor immune system.

Disadvantages of MatrigelCellular interactions with the ECM profoundly alter notonly gene expression pattern but also tumor cell behav-ior [105]. Tight ECM regulation in the tumor micro-environment is lost during engraftment, and tissuearchitecture degrades as PDX model tissue undergoespassages. Furthermore, Matrigel is frequently used to in-crease the engraftment rate in PDX models, since thepresence of growth factors in Matrigel can favor engraft-ment of primary tumor cells. However, it should be rec-ognized that this is a murine basement membraneextract [106], and Matrigel from murine sources can bea source of infectious murine viruses. In fact, treatmentof primary cancer cells with lactate dehydrogenase-

elevating virus-contaminated Matrigel in early studiesmay account for the reported instances of contamin-ation, and for this reason, some groups produced theirown Matrigel in order to avoid potential viral contamin-ation [107].

ConclusionsPDX models mimic not only the pathohistological andgenetic/epigenetic features of original tumor tissues butalso therapeutic responses to anti-cancer treatments.Therefore, PDX models have the potential to predict in-dividual responses to drugs and treatments, thereby fa-cilitating personalized medicine. Furthermore, many ofthe mechanisms underlying acquired drug resistance inmany tumor types have been elucidated using PDXmodels. Although the tumor stroma, including CAFs, isreplaced by mouse stroma during xenograft passages,the heterogeneity of the tumor microenvironment andthe metastatic potential of the tumor cells are main-tained in both subcutaneous and orthotopic PDXmodels. Notably, biofluorescence imaging of PDXmodels derived from organoids is a highly sensitivemethod for detecting micrometastatic lesions. Furtherinvestigations are necessary to develop strategies toevaluate the effects of immune-checkpoint inhibitors,because PDX models can only be established in im-munocompromised mouse strains.

AbbreviationsCAFs: Cancer-associated fibroblasts; CRC: Colorectal adenocarcinoma;CTC: Circulating tumor cell; ECM: Extracellular matrix; EMT: Epithelial-mesenchymal transition; GBM: Glioblastoma multiforme; GFP: Greenfluorescent protein; HGSOC: High-grade serous ovarian cancer;Hh: Hedgehog; IDH: Isocitrate dehydrogenase; mTOR: Mammalian target ofrapamycin; PDAC: Pancreatic ductal adenocarcinoma; PDXs: Patient-derivedxenografts; RTKs: Receptor tyrosine kinases; SREBP-1: Sterol regulatoryelement-binding protein-1; TNBC: Triple-negative breast cancer

AcknowledgementsI would like to thank Dr. Orimo A. of Juntendo University and Dr. MotoharaT. of Kumamoto University for advice and useful comments that helpedfinalize this review article.

Author’s contributionsGJY searched the literature and wrote the manuscript. The author read andapproved the final manuscript.

FundingThis review article was financially supported by the Japan Society for thePromotion of Science (19K23898).

Availability of data and materialsThe datasets used and/or analyzed during the current study are availablefrom the corresponding author on reasonable request.

Ethics approval and consent to participateNot applicable.

Consent for publicationGJY agrees to the content of this review paper and to being thecorresponding author.

Yoshida Journal of Hematology & Oncology (2020) 13:4 Page 13 of 16

Page 14: Applications of patient-derived tumor xenograft models and tumor … · 2020-01-07 · REVIEW Open Access Applications of patient-derived tumor xenograft models and tumor organoids

Competing interestsThere are no competing interests to be addressed.

Received: 17 October 2019 Accepted: 13 November 2019

References1. Kirschbaum A, Geisse NC, Sister TJ, Meyer LM. Effect of certain folic acid

antagonists on transplanted myeloid and lymphoid leukemias of the Fstrain of mice. Cancer Res. 1950;10(12):762–8.

2. Hutchinson L, Kirk R. High drug attrition rates--where are we going wrong?Nat Rev Clin Oncol. 2011;8(4):189–90.

3. Bertotti A, Migliardi G, Galimi F, Sassi F, Torti D, Isella C, Cora D, DiNicolantonio F, Buscarino M, Petti C, et al. A molecularly annotated platformof patient-derived xenografts (“xenopatients”) identifies HER2 as an effectivetherapeutic target in cetuximab-resistant colorectal cancer. Cancer Discov.2011;1(6):508–23.

4. DeRose YS, Wang G, Lin YC, Bernard PS, Buys SS, Ebbert MT, Factor R,Matsen C, Milash BA, Nelson E, et al. Tumor grafts derived from womenwith breast cancer authentically reflect tumor pathology, growth, metastasisand disease outcomes. Nat Med. 2011;17(11):1514–20.

5. Sivanand S, Pena-Llopis S, Zhao H, Kucejova B, Spence P, Pavia-Jimenez A,Yamasaki T, McBride DJ, Gillen J, Wolff NC et al: A validated tumorgraftmodel reveals activity of dovitinib against renal cell carcinoma. Sci TranslMed 2012, 4(137):137ra175.

6. Hidalgo M, Amant F, Biankin AV, Budinska E, Byrne AT, Caldas C, Clarke RB, de JongS, Jonkers J, Maelandsmo GM, et al. Patient-derived xenograft models: an emergingplatform for translational cancer research. Cancer Discov. 2014;4(9):998–1013.

7. Tentler JJ, Tan AC, Weekes CD, Jimeno A, Leong S, Pitts TM, Arcaroli JJ,Messersmith WA, Eckhardt SG. Patient-derived tumour xenografts as modelsfor oncology drug development. Nat Rev Clin Oncol. 2012;9(6):338–50.

8. Lawson DA, Bhakta NR, Kessenbrock K, Prummel KD, Yu Y, Takai K, Zhou A, EyobH, Balakrishnan S, Wang CY, et al. Single-cell analysis reveals a stem-cell programin human metastatic breast cancer cells. Nature. 2015;526(7571):131–5.

9. Xiao T, Li W, Wang X, Xu H, Yang J, Wu Q, Huang Y, Geradts J, Jiang P, Fei T,et al. Estrogen-regulated feedback loop limits the efficacy of estrogenreceptor-targeted breast cancer therapy. Proc Natl Acad Sci U S A. 2018;115(31):7869–78.

10. Reyal F, Guyader C, Decraene C, Lucchesi C, Auger N, Assayag F, De Plater L,Gentien D, Poupon MF, Cottu P, et al. Molecular profiling of patient-derivedbreast cancer xenografts. Breast Cancer Res. 2012;14(1):R11.

11. Zhao X, Liu Z, Yu L, Zhang Y, Baxter P, Voicu H, Gurusiddappa S, Luan J, SuJM, Leung HC, et al. Global gene expression profiling confirms themolecular fidelity of primary tumor-based orthotopic xenograft mousemodels of medulloblastoma. Neuro Oncol. 2012;14(5):574–83.

12. Misale S, Bozic I, Tong J, Peraza-Penton A, Lallo A, Baldi F, Lin KH, Truini M,Trusolino L, Bertotti A, et al. Vertical suppression of the EGFR pathway preventsonset of resistance in colorectal cancers. Nat Commun. 2015;6:8305.

13. Evans KW, Yuca E, Akcakanat A, Scott SM, Arango NP, Zheng X, Chen K,Tapia C, Tarco E, Eterovic AK, et al. A population of heterogeneous breastcancer patient-derived xenografts demonstrate broad activity of PARPinhibitor in BRCA1/2 wild-type tumors. Clin Cancer Res. 2017;23(21):6468–77.

14. Garcia PL, Miller AL, Gamblin TL, Council LN, Christein JD, Arnoletti JP, HeslinMJ, Reddy S, Richardson JH, Cui X, et al. JQ1 induces DNA damage andapoptosis, and inhibits tumor growth in a patient-derived xenograft modelof cholangiocarcinoma. Mol Cancer Ther. 2018;17(1):107–18.

15. Topp MD, Hartley L, Cook M, Heong V, Boehm E, McShane L, Pyman J,McNally O, Ananda S, Harrell M, et al. Molecular correlates of platinumresponse in human high-grade serous ovarian cancer patient-derivedxenografts. Mol Oncol. 2014;8(3):656–68.

16. Nunes M, Vrignaud P, Vacher S, Richon S, Lievre A, Cacheux W, Weiswald LB,Massonnet G, Chateau-Joubert S, Nicolas A, et al. Evaluating patient-derivedcolorectal cancer xenografts as preclinical models by comparison withpatient clinical data. Cancer Res. 2015;75(8):1560–6.

17. Fior R, Povoa V, Mendes RV, Carvalho T, Gomes A, Figueiredo N, Ferreira MG.Single-cell functional and chemosensitive profiling of combinatorialcolorectal therapy in zebrafish xenografts. Proc Natl Acad Sci U S A. 2017;114(39):E8234–43.

18. George E, Kim H, Krepler C, Wenz B, Makvandi M, Tanyi JL, Brown E, ZhangR, Brafford P, Jean S, et al. A patient-derived-xenograft platform to studyBRCA-deficient ovarian cancers. JCI Insight. 2017;2(1):e89760.

19. Sato T, Stange DE, Ferrante M, Vries RG, Van Es JH, Van den Brink S, VanHoudt WJ, Pronk A, Van Gorp J, Siersema PD, et al. Long-term expansion ofepithelial organoids from human colon, adenoma, adenocarcinoma, andBarrett's epithelium. Gastroenterology. 2011;141(5):1762–72.

20. Gao D, Vela I, Sboner A, Iaquinta PJ, Karthaus WR, Gopalan A, Dowling C,Wanjala JN, Undvall EA, Arora VK, et al. Organoid cultures derived frompatients with advanced prostate cancer. Cell. 2014;159(1):176–87.

21. Boj SF, Hwang CI, Baker LA, Chio II, Engle DD, Corbo V, Jager M, Ponz-Sarvise M, Tiriac H, Spector MS, et al. Organoid models of human andmouse ductal pancreatic cancer. Cell. 2015;160(1-2):324–38.

22. van de Wetering M, Francies HE, Francis JM, Bounova G, Iorio F, Pronk A,van Houdt W, van Gorp J, Taylor-Weiner A, Kester L, et al. Prospectivederivation of a living organoid biobank of colorectal cancer patients. Cell.2015;161(4):933–45.

23. Clevers H. Modeling development and disease with organoids. Cell. 2016;165(7):1586–97.

24. Fujii M, Shimokawa M, Date S, Takano A, Matano M, Nanki K, Ohta Y,Toshimitsu K, Nakazato Y, Kawasaki K, et al. A colorectal tumor organoidlibrary demonstrates progressive loss of niche factor requirements duringtumorigenesis. Cell Stem Cell. 2016;18(6):827–38.

25. Li M, Izpisua Belmonte JC. Organoids - preclinical models of human disease.N Engl J Med. 2019;380(6):569–79.

26. Smith RC, Tabar V. Constructing and deconstructing cancers using humanpluripotent stem cells and organoids. Cell Stem Cell. 2019;24(1):12–24.

27. Weeber F, van de Wetering M, Hoogstraat M, Dijkstra KK, Krijgsman O,Kuilman T, Gadellaa-van Hooijdonk CG, van der Velden DL, Peeper DS,Cuppen EP, et al. Preserved genetic diversity in organoids cultured frombiopsies of human colorectal cancer metastases. Proc Natl Acad Sci U S A.2015;112(43):13308–11.

28. Pauli C, Hopkins BD, Prandi D, Shaw R, Fedrizzi T, Sboner A, Sailer V, AugelloM, Puca L, Rosati R, et al. Personalized in vitro and in vivo cancer models toguide precision medicine. Cancer Discov. 2017;7(5):462–77.

29. Vlachogiannis G, Hedayat S, Vatsiou A, Jamin Y, Fernandez-Mateos J, Khan K,Lampis A, Eason K, Huntingford I, Burke R, et al. Patient-derived organoidsmodel treatment response of metastatic gastrointestinal cancers. Science.2018;359(6378):920–6.

30. Guo W, Giancotti FG. Integrin signalling during tumour progression. Nat RevMol Cell Biol. 2004;5(10):816–26.

31. Lu P, Takai K, Weaver VM, Werb Z: Extracellular matrix degradation andremodeling in development and disease. Cold Spring Harb Perspect Biol2011, 3(12).

32. Desgrosellier JS, Cheresh DA. Integrins in cancer: biological implications andtherapeutic opportunities. Nat Rev Cancer. 2010;10(1):9–22.

33. Hynes RO. Integrins: versatility, modulation, and signaling in cell adhesion.Cell. 1992;69(1):11–25.

34. Lelievre SA. Contributions of extracellular matrix signaling and tissuearchitecture to nuclear mechanisms and spatial organization of geneexpression control. Biochim Biophys Acta. 2009;1790(9):925–35.

35. Jinka R, Kapoor R, Sistla PG, Raj TA, Pande G. Alterations in cell-extracellular matrix interactions during progression of cancers. Int J CellBiol. 2012;2012:219196.

36. Yoshida GJ, Azuma A, Miura Y, Orimo A: Activated fibroblast programorchestrates tumor initiation and progression; molecular mechanisms andthe associated therapeutic strategies. Int J Mol Sci 2019, 20(9).

37. Mbeunkui F, Johann DJ Jr. Cancer and the tumor microenvironment: a reviewof an essential relationship. Cancer Chemother Pharmacol. 2009;63(4):571–82.

38. Runa F, Hamalian S, Meade K, Shisgal P, Gray PC, Kelber JA. Tumormicroenvironment heterogeneity: challenges and opportunities. Curr MolBiol Rep. 2017;3(4):218–29.

39. Foster DS, Jones RE, Ransom RC, Longaker MT, Norton JA: The evolvingrelationship of wound healing and tumor stroma. JCI Insight 2018, 3(18).

40. LeBleu VS, Kalluri R: A peek into cancer-associated fibroblasts: origins,functions and translational impact. Dis Model Mech 2018, 11(4).

41. Bachman KE, Park BH. Duel nature of TGF-beta signaling: tumor suppressorvs. tumor promoter. Curr Opin Oncol. 2005;17(1):49–54.

42. Batlle E, Massague J. Transforming growth factor-beta signaling in immunityand cancer. Immunity. 2019;50(4):924–40.

43. Stanisavljevic J, Loubat-Casanovas J, Herrera M, Luque T, Pena R, Lluch A,Albanell J, Bonilla F, Rovira A, Pena C, et al. Snail1-expressing fibroblasts inthe tumor microenvironment display mechanical properties that supportmetastasis. Cancer Res. 2015;75(2):284–95.

Yoshida Journal of Hematology & Oncology (2020) 13:4 Page 14 of 16

Page 15: Applications of patient-derived tumor xenograft models and tumor … · 2020-01-07 · REVIEW Open Access Applications of patient-derived tumor xenograft models and tumor organoids

44. Pompili L, Porru M, Caruso C, Biroccio A, Leonetti C. Patient-derivedxenografts: a relevant preclinical model for drug development. J Exp ClinCancer Res. 2016;35(1):189.

45. Mosmann TR, Yokota T, Kastelein R, Zurawski SM, Arai N, Takebe Y. Species-specificity of T cell stimulating activities of IL 2 and BSF-1 (IL 4): comparisonof normal and recombinant, mouse and human IL 2 and BSF-1 (IL 4). JImmunol. 1987;138(6):1813–6.

46. Collins MK. Species specificity of interleukin 2 binding to individual receptorcomponents. Eur J Immunol. 1989;19(8):1517–20.

47. Eisenman J, Ahdieh M, Beers C, Brasel K, Kennedy MK, Le T, Bonnert TP,Paxton RJ, Park LS. Interleukin-15 interactions with interleukin-15receptor complexes: characterization and species specificity. Cytokine.2002;20(3):121–9.

48. Huntington ND, Legrand N, Alves NL, Jaron B, Weijer K, Plet A, CorcuffE, Mortier E, Jacques Y, Spits H, et al. IL-15 trans-presentation promoteshuman NK cell development and differentiation in vivo. J Exp Med.2009;206(1):25–34.

49. Kondrashova O, Topp M, Nesic K, Lieschke E, Ho GY, Harrell MI, ZapparoliGV, Hadley A, Holian R, Boehm E, et al. Methylation of all BRCA1 copiespredicts response to the PARP inhibitor rucaparib in ovarian carcinoma. NatCommun. 2018;9(1):3970.

50. Lee SH, Hu W, Matulay JT, Silva MV, Owczarek TB, Kim K, Chua CW, BarlowLJ, Kandoth C, Williams AB, et al. Tumor evolution and drug response inpatient-derived organoid models of bladder cancer. Cell. 2018;173(2):515–28e517.

51. Lancaster MA, Knoblich JA. Organogenesis in a dish: modelingdevelopment and disease using organoid technologies. Science. 2014;345(6194):1247125.

52. Clohessy JG, Pandolfi PP. Mouse hospital and co-clinical trial project--frombench to bedside. Nat Rev Clin Oncol. 2015;12(8):491–8.

53. Ben-David U, Ha G, Tseng YY, Greenwald NF, Oh C, Shih J, McFarland JM,Wong B, Boehm JS, Beroukhim R, et al. Patient-derived xenografts undergomouse-specific tumor evolution. Nat Genet. 2017;49(11):1567–75.

54. Camidge DR, Pao W, Sequist LV. Acquired resistance to TKIs in solidtumours: learning from lung cancer. Nat Rev Clin Oncol. 2014;11(8):473–81.

55. Restifo NP, Smyth MJ, Snyder A. Acquired resistance to immunotherapy andfuture challenges. Nat Rev Cancer. 2016;16(2):121–6.

56. Okazawa Y, Mizukoshi K, Koyama Y, Okubo S, Komiyama H, Kojima Y, GotoM, Habu S, Hino O, Sakamoto K, et al. High-sensitivity Detection ofMicrometastases Generated by GFP Lentivirus-transduced OrganoidsCultured from a Patient-derived Colon Tumor. J Vis Exp. 2018;136.

57. Kinsey CG, Camolotto SA, Boespflug AM, Guillen KP, Foth M, Truong A,Schuman SS, Shea JE, Seipp MT, Yap JT, et al. Protective autophagy elicitedby RAF-->MEK-->ERK inhibition suggests a treatment strategy for RAS-drivencancers. Nat Med. 2019;25(4):620–7.

58. Yoshida GJ. Therapeutic strategies of drug repositioning targetingautophagy to induce cancer cell death: from pathophysiology to treatment.J Hematol Oncol. 2017;10(1):67.

59. Singh DK, Kollipara RK, Vemireddy V, Yang XL, Sun Y, Regmi N, Klingler S,Hatanpaa KJ, Raisanen J, Cho SK, et al. Oncogenes activate an autonomoustranscriptional regulatory circuit that drives glioblastoma. Cell Rep. 2017;18(4):961–76.

60. Tornin J, Martinez-Cruzado L, Santos L, Rodriguez A, Nunez LE, Oro P,Hermosilla MA, Allonca E, Fernandez-Garcia MT, Astudillo A, et al. Inhibitionof SP1 by the mithramycin analog EC-8042 efficiently targets tumorinitiating cells in sarcoma. Oncotarget. 2016;7(21):30935–50.

61. Vanner RJ, Remke M, Gallo M, Selvadurai HJ, Coutinho F, Lee L, Kushida M,Head R, Morrissy S, Zhu X, et al. Quiescent sox2(+) cells drive hierarchicalgrowth and relapse in sonic hedgehog subgroup medulloblastoma. CancerCell. 2014;26(1):33–47.

62. Chen S, Giannakou A, Wyman S, Gruzas J, Golas J, Zhong W, Loreth C,Sridharan L, Yamin TT, Damelin M, et al. Cancer-associated fibroblastssuppress SOX2-induced dysplasia in a lung squamous cancer coculture.Proc Natl Acad Sci U S A. 2018;115(50):E11671–80.

63. Kalluri R, Weinberg RA. The basics of epithelial-mesenchymal transition. JClin Invest. 2009;119(6):1420–8.

64. Bertolini G, D'Amico L, Moro M, Landoni E, Perego P, Miceli R, Gatti L,Andriani F, Wong D, Caserini R, et al. Microenvironment-modulatedmetastatic CD133+/CXCR4+/EpCAM- lung cancer-initiating cells sustaintumor dissemination and correlate with poor prognosis. Cancer Res. 2015;75(17):3636–49.

65. Saitoh M. Involvement of partial EMT in cancer progression. J Biochem.2018;164(4):257–64.

66. Wang X, Enomoto A, Asai N, Kato T, Takahashi M. Collective invasion of cancer:Perspectives from pathology and development. Pathol Int. 2016;66(4):183–92.

67. Ferone G, Song JY, Sutherland KD, Bhaskaran R, Monkhorst K, Lambooij JP,Proost N, Gargiulo G, Berns A. SOX2 is the determining oncogenic switch inpromoting lung squamous cell carcinoma from different cells of origin.Cancer Cell. 2016;30(4):519–32.

68. Watanabe H, Ma Q, Peng S, Adelmant G, Swain D, Song W, Fox C, FrancisJM, Pedamallu CS, DeLuca DS, et al. SOX2 and p63 colocalize at genetic lociin squamous cell carcinomas. J Clin Invest. 2014;124(4):1636–45.

69. Murray NR, Justilien V, Fields AP. SOX2 determines lineage restriction: modelinglung squamous cell carcinoma in the mouse. Cancer Cell. 2016;30(4):505–7.

70. Luga V, Zhang L, Viloria-Petit AM, Ogunjimi AA, Inanlou MR, Chiu E,Buchanan M, Hosein AN, Basik M, Wrana JL. Exosomes mediate stromalmobilization of autocrine Wnt-PCP signaling in breast cancer cell migration.Cell. 2012;151(7):1542–56.

71. Talebi A, Dehairs J, Rambow F, Rogiers A, Nittner D, Derua R,Vanderhoydonc F, Duarte JAG, Bosisio F, Van den Eynde K, et al. SustainedSREBP-1-dependent lipogenesis as a key mediator of resistance to BRAF-targeted therapy. Nat Commun. 2018;9(1):2500.

72. Dewaele M, Tabaglio T, Willekens K, Bezzi M, Teo SX, Low DH, Koh CM,Rambow F, Fiers M, Rogiers A, et al. Antisense oligonucleotide-mediated MDM4 exon 6 skipping impairs tumor growth. J Clin Invest.2016;126(1):68–84.

73. Choi CH, Ryu JY, Cho YJ, Jeon HK, Choi JJ, Ylaya K, Lee YY, Kim TJ, Chung JY,Hewitt SM, et al. The anti-cancer effects of itraconazole in epithelial ovariancancer. Sci Rep. 2017;7(1):6552.

74. Kim J, Tang JY, Gong R, Kim J, Lee JJ, Clemons KV, Chong CR, Chang KS,Fereshteh M, Gardner D, et al. Itraconazole, a commonly used antifungalthat inhibits Hedgehog pathway activity and cancer growth. Cancer Cell.2010;17(4):388–99.

75. Yoshida GJ. Metabolic reprogramming: the emerging concept andassociated therapeutic strategies. J Exp Clin Cancer Res. 2015;34:111.

76. Zhou H, Qian W, Uckun FM, Wang L, Wang YA, Chen H, Kooby D, Yu Q,Lipowska M, Staley CA, et al. IGF1 receptor targeted theranosticnanoparticles for targeted and image-guided therapy of pancreatic cancer.ACS Nano. 2015;9(8):7976–91.

77. Witkiewicz AK, Balaji U, Eslinger C, McMillan E, Conway W, Posner B, MillsGB, O'Reilly EM, Knudsen ES. Integrated patient-derived models delineateindividualized therapeutic vulnerabilities of pancreatic cancer. Cell Rep.2016;16(7):2017–31.

78. Pettazzoni P, Viale A, Shah P, Carugo A, Ying H, Wang H, Genovese G, SethS, Minelli R, Green T, et al. Genetic events that limit the efficacy of MEK andRTK inhibitor therapies in a mouse model of KRAS-driven pancreatic cancer.Cancer Res. 2015;75(6):1091–101.

79. Rajeshkumar NV, Yabuuchi S, Pai SG, De Oliveira E, Kamphorst JJ,Rabinowitz JD, Tejero H, Al-Shahrour F, Hidalgo M, Maitra A, et al.Treatment of pancreatic cancer patient-derived xenograft panel withmetabolic inhibitors reveals efficacy of phenformin. Clin Cancer Res.2017;23(18):5639–47.

80. Bailey CJ, Turner RC. Metformin. N Engl J Med. 1996;334(9):574–9.81. Zou S, Li J, Zhou H, Frech C, Jiang X, Chu JS, Zhao X, Li Y, Li Q, Wang H,

et al. Mutational landscape of intrahepatic cholangiocarcinoma. NatCommun. 2014;5:5696.

82. Lee G, Auffinger B, Guo D, Hasan T, Deheeger M, Tobias AL, Kim JY, AtashiF, Zhang L, Lesniak MS, et al. Dedifferentiation of glioma cells to gliomastem-like cells by therapeutic stress-induced HIF signaling in the recurrentGBM model. Mol Cancer Ther. 2016;15(12):3064–76.

83. Fack F, Tardito S, Hochart G, Oudin A, Zheng L, Fritah S, Golebiewska A,Nazarov PV, Bernard A, Hau AC, et al. Altered metabolic landscape in IDH-mutant gliomas affects phospholipid, energy, and oxidative stress pathways.EMBO Mol Med. 2017;9(12):1681–95.

84. Garner EF, Williams AP, Stafman LL, Aye JM, Mroczek-Musulman E, Moore BP,Stewart JE, Friedman GK, Beierle EA. FTY720 decreases tumorigenesis in group3 medulloblastoma patient-derived xenografts. Sci Rep. 2018;8(1):6913.

85. Chun J, Hartung HP. Mechanism of action of oral fingolimod (FTY720) inmultiple sclerosis. Clin Neuropharmacol. 2010;33(2):91–101.

86. Goga A, Yang D, Tward AD, Morgan DO, Bishop JM. Inhibition of CDK1as a potential therapy for tumors over-expressing MYC. Nat Med. 2007;13(7):820–7.

Yoshida Journal of Hematology & Oncology (2020) 13:4 Page 15 of 16

Page 16: Applications of patient-derived tumor xenograft models and tumor … · 2020-01-07 · REVIEW Open Access Applications of patient-derived tumor xenograft models and tumor organoids

87. Horiuchi D, Kusdra L, Huskey NE, Chandriani S, Lenburg ME, Gonzalez-Angulo AM, Creasman KJ, Bazarov AV, Smyth JW, Davis SE, et al. MYCpathway activation in triple-negative breast cancer is synthetic lethal withCDK inhibition. J Exp Med. 2012;209(4):679–96.

88. Yu J, Qin B, Moyer AM, Nowsheen S, Liu T, Qin S, Zhuang Y, Liu D, Lu SW,Kalari KR, et al. DNA methyltransferase expression in triple-negative breastcancer predicts sensitivity to decitabine. J Clin Invest. 2018;128(6):2376–88.

89. El Ayachi I, Fatima I, Wend P, Alva-Ornelas JA, Runke S, Kuenzinger WL, SilvaJ, Silva W, Gray JK, Lehr S, et al. The WNT10B network is associated withsurvival and metastases in chemoresistant triple-negative breast cancer.Cancer Res. 2019;79(5):982–93.

90. Weeden CE, Holik AZ, Young RJ, Ma SB, Garnier JM, Fox SB, Antippa P, IrvingLB, Steinfort DP, Wright GM, et al. Cisplatin increases sensitivity to FGFRinhibition in patient-derived xenograft models of lung squamous cellcarcinoma. Mol Cancer Ther. 2017;16(8):1610–22.

91. Drapkin BJ, George J, Christensen CL, Mino-Kenudson M, Dries R,Sundaresan T, Phat S, Myers DT, Zhong J, Igo P, et al. Genomic andfunctional fidelity of small cell lung cancer patient-derived xenografts.Cancer Discov. 2018;8(5):600–15.

92. Mollaoglu G, Guthrie MR, Bohm S, Bragelmann J, Can I, Ballieu PM, Marx A,George J, Heinen C, Chalishazar MD, et al. MYC drives progression of smallcell lung cancer to a variant neuroendocrine subtype with vulnerability toaurora kinase inhibition. Cancer Cell. 2017;31(2):270–85.

93. Yoshida GJ. Emerging roles of Myc in stem cell biology and novel tumortherapies. J Exp Clin Cancer Res. 2018;37(1):173.

94. Gong K, Guo G, Gerber DE, Gao B, Peyton M, Huang C, Minna JD, Hatanpaa KJ,Kernstine K, Cai L, et al. TNF-driven adaptive response mediates resistance toEGFR inhibition in lung cancer. J Clin Invest. 2018;128(6):2500–18.

95. Hirata E, Girotti MR, Viros A, Hooper S, Spencer-Dene B, Matsuda M, Larkin J,Marais R, Sahai E. Intravital imaging reveals how BRAF inhibition generatesdrug-tolerant microenvironments with high integrin beta1/FAK signaling.Cancer Cell. 2015;27(4):574–88.

96. Krepler C, Sproesser K, Brafford P, Beqiri M, Garman B, Xiao M, Shannan B,Watters A, Perego M, Zhang G, et al. A comprehensive patient-derivedxenograft collection representing the heterogeneity of melanoma. Cell Rep.2017;21(7):1953–67.

97. Einarsdottir BO, Karlsson J, Soderberg EMV, Lindberg MF, Funck-Brentano E,Jespersen H, Brynjolfsson SF, Bagge RO, Carstam L, Scobie M, et al. Apatient-derived xenograft pre-clinical trial reveals treatment responses and aresistance mechanism to karonudib in metastatic melanoma. Cell Death Dis.2018;9(8):810.

98. Warpman Berglund U, Sanjiv K, Gad H, Kalderen C, Koolmeister T, Pham T,Gokturk C, Jafari R, Maddalo G, Seashore-Ludlow B, et al. Validation anddevelopment of MTH1 inhibitors for treatment of cancer. Ann Oncol. 2016;27(12):2275–83.

99. Grasset EM, Bertero T, Bozec A, Friard J, Bourget I, Pisano S, Lecacheur M,Maiel M, Bailleux C, Emelyanov A, et al. Matrix stiffening and EGFRcooperate to promote the collective invasion of cancer cells. Cancer Res.2018;78(18):5229–42.

100. Liu JF, Palakurthi S, Zeng Q, Zhou S, Ivanova E, Huang W, Zervantonakis IK,Selfors LM, Shen Y, Pritchard CC, et al. Establishment of patient-derivedtumor xenograft models of epithelial ovarian cancer for preclinicalevaluation of novel therapeutics. Clin Cancer Res. 2017;23(5):1263–73.

101. Kim H, George E, Ragland R, Rafial S, Zhang R, Krepler C, Morgan M, HerlynM, Brown E, Simpkins F. Targeting the ATR/CHK1 axis with PARP inhibitionresults in tumor regression in BRCA-mutant ovarian cancer models. ClinCancer Res. 2017;23(12):3097–108.

102. Cancer Genome Atlas Research N. Integrated genomic analyses of ovariancarcinoma. Nature. 2011;474(7353):609–15.

103. Li S, Shen D, Shao J, Crowder R, Liu W, Prat A, He X, Liu S, Hoog J, Lu C,et al. Endocrine-therapy-resistant ESR1 variants revealed by genomiccharacterization of breast-cancer-derived xenografts. Cell Rep. 2013;4(6):1116–30.

104. Junttila MR, de Sauvage FJ. Influence of tumour micro-environmentheterogeneity on therapeutic response. Nature. 2013;501(7467):346–54.

105. Wang CC, Bajikar SS, Jamal L, Atkins KA, Janes KA. A time- and matrix-dependent TGFBR3-JUND-KRT5 regulatory circuit in single breast epithelialcells and basal-like premalignancies. Nat Cell Biol. 2014;16(4):345–56.

106. Chou JL, Shen ZX, Stolfi RL, Martin DS, Waxman S. Effects of extracellularmatrix on the growth and casein gene expression of primary mousemammary tumor cells in vitro. Cancer Res. 1989;49(19):5371–6.

107. DeRose YS, Gligorich KM, Wang G, Georgelas A, Bowman P, Courdy SJ,Welm AL, Welm BE: Patient-derived models of human breast cancer:protocols for in vitro and in vivo applications in tumor biology andtranslational medicine. Curr Protoc Pharmacol 2013, Chapter 14:Unit14 23.

Publisher’s NoteSpringer Nature remains neutral with regard to jurisdictional claims inpublished maps and institutional affiliations.

Yoshida Journal of Hematology & Oncology (2020) 13:4 Page 16 of 16