Abstract
Colorectal cancer (CRC) is one of the most popular malignancies globally, with 930 000 deaths in 2020. The evaluation of CRC‐related pathogenesis and the discovery of potential therapeutic targets will be meaningful and helpful for improving CRC treatment. With huge efforts made in past decades, the systematic treatment regimens have been applied to improve the prognosis of CRC patients. However, the sensitivity of CRC to chemotherapy and targeted therapy is different from person to person, which is an important cause of treatment failure. The emergence of patient‐derived xenograft (PDX) models shows great potential to alleviate the straits. PDX models possess similar genetic and pathological characteristics as the features of primary tumors. Moreover, PDX has the ability to mimic the tumor microenvironment of the original tumor. Thus, the PDX model is an important tool to screen precise drugs for individualized treatment, seek predictive biomarkers for prognosis supervision, and evaluate the unknown mechanism in basic research. This paper reviews the recent advances in constructed methods and applications of the CRC PDX model, aiming to provide new knowledge for CRC basic research and therapeutics.
Keywords: colorectal cancer, drug discovery, patient‐derived xenograft model, precision medicine, tumor microenvironment
The establishment of PDX bio‐bank for precision medicine. Specimens from CRC patients enrolled in the study are submitted to PDX bio‐bank. Researchers can take advantages of the bio‐banks to screen drugs, uncover biomarkers, and study the basic mechanisms. The latent drugs will submit to animal tests using PDX in bio‐banks. The effective drugs and biomarkers will improve therapeutic effects.

1. BACKGROUND
Colorectal cancer (CRC), one of the most deadly cancers, is the fourth most common cause of cancer‐related deaths. 1 Currently, surgery is the main treatment option for primary and metastatic CRC, and chemotherapy and targeted drugs are regularly used as adjuvant regimens. 2 , 3 , 4 Despite improvements in screening and treatment, the number of individuals newly diagnosed is increasing rapidly. Moreover, metastasis, recurrence, and chemoresistance are common in CRC patients after treatment, which leads to the dismal prognosis and high mortality of CRC patients. 5 , 6 , 7 These issues lead to the need for more advancements in CRC treatment.
About 5% of CRCs are hereditary tumor syndromes, such as familial adenomatous polyposis (FAP) and Lynch syndrome. 8 Most of the CRCs are sporadic as the comprehensive effects of various factors, including somatic genetic alterations, chronic inflammation, lifestyle, and environmental stimuli. 9 These genetic and nongenetic risk factors work together to promote CRC development and progression. 10 , 11 , 12 , 13 To date, the mechanisms involved in the interactions of these factors driving CRC development and progression are not fully determined. More important, CRC is a significantly heterogeneous disease with distinct variations in molecular features, which cause personalized response to therapy. 14 , 15 Tumor microenvironment (TME), another reason for tumor heterogeneity, has been found to be closely associated with individual tumor progression. 16 TME plays functional and structural roles in tumor cell growth and proliferation. 16 , 17 TME exhibits the ability to limit the efficacy of therapeutic agents, modulate tumor metastasis, and prevent recurrence. 18 , 19 Therefore, TME is a fertile ground for the development of new treatments. 20 , 21 TME possesses a dynamic composition, including various cell types and extracellular matrix, such as cancer‐associated fibroblasts (CAFs), regulatory T cells (Tregs), tumor‐associated macrophages, and myeloid‐derived suppressor cells, which coordinate with one another to promote tumor progression. 22
The issues of heterogeneity suggest that personalized treatment is required for improving the prognosis of CRC patients based on the individual characteristics of cancer biology. 23 The mutations of some crucial genes, such as KRAS, BRAF, and microsatellite, have been used as important markers for drug selection. 24 Recently, strategies targeting TME have been developed and applied in the treatment of human cancers, including CRC. Daclizumab, a monoclonal antibody against the CD25 receptor, has been approved by the U.S. Food and Drug Administration and has the ability to effectively decrease circulating Tregs. 25 The combination of oxaliplatin and programmed death ligand‐1 therapy to inhibit TME has been reported to be effective in CRC patients with microsatellite instability. 26
2. TYPICAL CRC PRECLINICAL MODELS
Mice are frequently used in CRC research and drug screening. Currently, mouse models are divided into main two groups, primary tumor model and transplanted tumor model. The primary tumor models refer to spontaneous CRCs in mice or CRCs induced by genetic engineering or chemical reagents in mice. 27 CRC transplantation models are built by transplanting human or mouse primary CRC tissues or cells into mice. 10 , 28 Based on graft sources, the model can be divided into allografts and xenografts. Mouse‐derived CRC tissues or cells, such as MC38 mouse colon cancer cell line, are used in the construction of allografts. 29 Generally, xenograft models are established by transplanting human CRC tissues or cells into mice. Transplanted CRC models present important advantages for screening drugs, evaluating drug effect, or predicting clinical effects of drugs. 30 The primary tumor models are more suitable to evaluate the underlying mechanisms of CRC development or metastasis. The features and applications of these models are summarized in Figure 1.
FIGURE 1.

The frequently used mouse models in CRC (colorectal cancer) research. The features and applications of mouse models, including AOM (azoxymethane)/DSS (dextran sulfate sodium)–induced model, spontaneous CRC, GEMMs (genetically engineered mouse models), CDX (cell line‐derived xenograft), and PDX (patient‐derived xenograft) models, are summarized
A small number of old mice develop tumor, including CRC, under natural conditions. However, it is inconvenient to establish these models because of low success rate and more time consumption. Gene mutations using genetic engineering or chemical reagents contribute to an increase in the tumor incidence in mice to fit the needs of experiments. 31 , 32 Thus, these models have been widely used in CRC research. The genetically engineered mouse models (GEMMs) carrying the mutation in Apc gene have been found to suffer from spontaneously multiple intestinal neoplasia similar to patients with FAP, which were used to study the molecular mechanisms involved in the early stage of CRC. 33 Subsequently, transgenic mice with alternative Apc mutations and/or in combination with other mutations of oncogene or tumor suppressor gene such as Ras, Pten, and Braf were established for further understanding of tumor biology. 11 , 34 The frequently used GEMMs are presented in Table 1. GEMMs with accurate gene mutations are helpful for evaluating the functions of specific genes as well as for evaluating the related molecular pathway of tumorigenesis. As the cost of GEMMs decreases, they are frequently applied in studying the pathogenesis of CRC. However, these models have limitations due to the great decrease in life span in mice with multiple mutations.
TABLE 1.
The development of murine intestinal GEMMs
| Years | Target genes | Related disease in humans |
|---|---|---|
| 1990 | Apc min/+ | Recapitulates the disease observed in FAP patients |
| 1997 | Apc 580S/580S | Colonic adenomas |
| 2000+ | Apc 580S | Invasive adenocarcinomas but not metastasis |
| Apc, Kras G12V | ||
| Apc, Pten−/− | ||
| Apc, Pik3 CA | ||
| Apc, TGFBR1/2/−/− | ||
| 2005+ | Kras, Braf mutation | Adenocarcinoma that had the capacity to metastasize |
Abbreviations: FAP, familial adenomatous polyposis; GEMMs, genetically engineered mouse models.
Alternatively, the carcinogenic‐induced model was developed for CRC research. The mouse models are established by injecting azoxymethane combined with dextran sulfate sodium in a time‐ and dose‐specific manner to mimic the aberrant crypt foci‐adenoma‐carcinoma progress, which occurs in CRC patients. 35 , 36 , 37 Compared with GEMMs, this model is much easier, more convenient, less time consuming, and inexpensive for researchers. Therefore, carcinogenic‐induced CRC model is one of the commonly used models in CRC research and drug screening. Although GEMMs and carcinogenic‐induced models are important tools in basic research and drug discovery of CRC, they still fail to remodel some critical features of clinical trials. 38 These models lack genetic consistency, a diversified diet, varied lifestyle, and an equal microbiome similar to human CRCs, all of which affect the disease's onset and course. 39 , 40 Moreover, the absence of inter‐tumoral heterogeneity because of poor genetic heterogeneity in inbred mice compared to outbred humans also decreases the potential of results in translational clinical research. These differences urge us to develop new models that more exactly display the characteristics of human CRCs.
Human cancer cell lines are typical in vitro model systems commonly applied in drug discovery and basic research. 41 , 42 , 43 , 44 More than 100 cell lines have been established as CRC cell lines from cell line banks worldwide. Cell line–derived xenograft (CDX) models developed by implanting cancer cell lines into immunodeficient mice contribute to the discovery of underlying mechanisms and the development of cancer drugs. 10 , 45 , 46 The most common CRC cell lines used for establishing the CRC CDX model are presented in Table 2. The CDX model is very popular in CRC research and drug discovery globally because it consumes less time, produces high yield, and is inexpensive. However, this traditional model fails to simulate tumor genetic heterogeneity of the primary tumor because of losing original inheritance and missing relevant components of TME during in vitro passage. 47 Moreover, the artificial culture conditions may cause alterations at genetic and epigenetic levels during serial passaging with enrichment for specific subclones. 48 Recently, the U.S. National Cancer Institute (NCI) has deprioritized the NCI‐60 human cancer cell lines for most drug screening processes. 49 Currently, patient‐derived xenograft (PDX) models have been established to overcome the limitations of the CDX model to improve our understanding of tumor biology and to better identify novel drugs for cancer treatment. 50 , 51 , 52
TABLE 2.
Frequently used cell lines in CRC research
| Name | Source | Gene signature | Tumorigenicity in nude mice |
|---|---|---|---|
| HCT116 | Colon cancer | CEA (+) | Yes |
| SW480 | Rectum adenocarcinoma in situ | c‐myc (+), K‐ras (+), H‐ras (+), N‐ras (+), myb (+), sis (+), fos (+), TP53 mutation | Yes |
| SW620 | Lymph node metastasis of rectum adenocarcinoma | Similar to SW480 | Yes |
| HCT‐15 | Colon cancer | CSAp (−) | |
| SW1116 | Colon cancer | c‐myc (+), K‐ras (+), H‐ras (+), myb (+), sis(+), fos (+) | Yes |
| LOVO | Metastatic colon cancer | c‐myc (+), K‐ras (+), H‐ras (+), N‐ras (+), myb (+), sis (+), fos (+) | Yes |
| HT‐29 | Colon cancer | c‐myc (+), K‐ras (+), H‐ras (+), N‐ras (+), myb (+), sis (+), fos (+), TP53 mutation | Yes |
| LS180 | Colon cancer | CEA (+) | Yes |
| LS174T | Colon cancer | c‐myc (+), K‐ras (+), H‐ras (+), N‐ras (+), myb (+), fos (+) | Yes |
| NCI‐H716 | Colorectal cancer | CEA (−) | Yes |
| Caco‐2 | Rectum cancer | Keratoprotein (+) | Yes |
| COLO205 | Colon cancer | CEA (+) | Yes |
| DLD1 | Colorectal cancer | c‐myc (+), K‐ras (+), H‐ras (+), N‐ras (+), myb (+), fos (+), CSAp (−) | Yes |
| RKO | Colon cancer | Wild‐type TP53 | Yes |
Abbreviations: CEA, carcinoembryonic antigen; CRC, colorectal cancer; CSAp: colon‐specific antigen.
3. DEVELOPMENT OF PDX MODELS
In an attempt to mimic the original tumor inter‐ and intra‐heterogeneity, the PDX model has been developed by directly implanting tumor tissues from patients into immunodeficient animals. 53 , 54 , 55 In the 1950 s, the first case of tumor xenograft in mice was successfully established. Currently, the process of establishing PDXs in mice from primary or metastatic CRCs has been widely reported. 53 , 56 In the early stage, it was difficult to establish the CRC PDX model in most laboratories due to low success rate, complicated procedure, and high cost. With constant efforts, the simplified and standardized procedure for CRC PDX has been established, and PDX models have been widely applied in CRC research. 57 , 58 The timeline of PDX model development is shown in Figure 2. This model preserves the parental tumor construction and the existing communications between tumor cells and stromal and immune cells. 59 , 60 Many studies have confirmed the high consistency between PDX and the corresponding primary tumor tissues in histopathological and molecular characteristics even after several passages, indicating that PDX models keep genetic stability from the primary tumors. 61 , 62 , 63 , 64 The results from the consensus molecular subtype (CMS) classification of PDXs and the matched primary CRCs have indicated that no variation in CMS frequencies was found between the two groups. 65 , 66 The evaluation of carcinoembryonic antigen and cytokeratin also supports that PDX maintains histopathological features of the original human cancer tissues. 67 , 68
FIGURE 2.

The timeline of PDX (patient‐derived xenograft) model development. NOD‐SCID, nonobese diabetic/severe combined immunodeficiency; NOG, NOD.Cg‐Prkdc (scid) II2rg (tm1Sug)/JicCrl; NSG, NOD‐SCID IL2rg−/−; CIEA, Central Institute for Experimental Animals
Currently, standardized procedures have been constructed for the production, quality assurance, and application of CRC PDX (Figure 3). 69 Generally, tumor tissues, obtained by surgery or biopsy procedures, are directly engrafted as pieces (~30 mm3) after resection or after cryopreservation. 70 Optionally, tumor tissues can be pretreated in Matrigel (a solubilized basement membrane matrix) or combined with human fibroblasts before engraftment to improve implantation achievement. PDX models are developed either heterotopically, through subcutaneous implantation into the dorsal area of mice, or orthotopically, by direct implantation into the anatomical site of origin. For CRC, subcutaneous implantation is the most commonly used procedure to establish PDXs, because it is easy to operate, monitor, and resect with good tumor engraftment. The orthoxenografts are helpful to evaluate local invasive growth of primary tumors, study tumor–host communications in their anatomical context, and observe the site‐specific dependence on therapy. 71 , 72 Given that microsurgical skills and small animal imaging are required for generating and monitoring orthoxenografts, this method is more difficult as a preclinical model compared with heterotopic xenograft. The median overall success rate for constructing CRC PDX is 70%, and it often takes 2–4 months. 11 , 73 These parameters may vary based on sample type, tumor stage, and recipient strain. It is reported that epithelial subtypes displayed lower engraftment rate than microsatellite instability tumors. 66 The engraftment success rate from metastatic samples is higher than that from primary tumors, indicating that the success is affected by tumor stage. 66 , 74 In terms of recipient strain, immune‐deficient mice such as NOD/SCID (nonobese diabetic/severe combined immunodeficiency) and NOD/SCID/IL2γ‐receptor null (NSG) are the most suitable hosts for PDX generation because of their lower immune rejection and higher engraftment rate. 53 , 75 , 76 Alternatively, it is reported that zebrafish can be used for constructing the PDX model. 77 , 78 , 79
FIGURE 3.

The establishment, storage, and application of CRC (colorectal cancer) PDX (patient‐derived xenograft). The tumor tissues derived from patients are implanted into mice with immunodeficiency. The model can be used in drug testing, basic research, and biomarker discovery
4. THE APPLICATION OF PDX IN CRC RESEARCH
PDX is a promising model for addressing clinically relevant issues such as drug screening, prognosis supervision, discovery of biomarkers, and evaluation of the involved mechanisms.
4.1. Drug discovery
It has been found that both the histology and expression of tumor‐associated markers (e.g., epithelial cell adhesion molecule, E‐cadherin, carcinoembryonic antigen, and Ki67) can be maintained during passage in nude mice. 80 Importantly, the PDX model also displays a good response to multiple chemotherapeutic agents, consistent with the response in patients, indicating that this model can be used for drug screening. 64 The establishment of large PDX cohorts originating from the same tumor sample helps analyze genotype‐response correlations and identify optimal drugs in patients. 81 Thus, the application of the PDX model provides more evidence for the potential drug before clinical studies start. Now, more studies employ the PDX model to uncover potential drugs for CRC treatment (Table 3).
TABLE 3.
Drug discovery in CRC research using PDX models
| Drugs | Function | Reference |
|---|---|---|
| Interferon‐α | Enhances the antitumor effects of 5‐fluorouracil | Ref. 84 |
| XMU‐MP‐2 | Attenuates chemoresistance of CRC | Ref. 85 |
| G6PD shRNA | Increases the antitumor effects of oxaliplatin | Ref. 86 |
| Dasatinib | Sensitizes liver metastatic CRC to oxaliplatin | Refs. 87, 88 |
| Anti‐TIMP‐2 antibody, U0126 | Attenuates the drug resistance of CRC cells to 5‐fluorouracil | Ref. 89 |
| AZ31 | Enhances the antitumor effects of irinotecan | Ref. 90 |
| GC1118 | Exhibits therapeutic efficacy in KRAS mutation‐driven PDX | Ref. 94 |
| TAK‐960 | Displays active antiproliferative effect against CRC carrying KRAS mutation | Ref. 95 |
| ABT‐263, NCB‐0846 | Exhibits antitumor effects in KRAS/BRAF‐mutated CRC PDX | Ref. 97 |
| Bortezomib, everolimus | Exhibits antitumor effects in KRAS‐mutated CRC | Ref. 98 |
| RGX‐202 | Suppresses CRC growth in KRAS‐mutated CRC PDX | Ref. 99 |
| Volitinib, apatinib | Increases apoptosis in c‐Met overexpressing CRC | Ref. 101 |
| ENMD‐2076 | Exhibits a promising antitumor activity in CRC PDX | Ref. 102 |
| SN38‐loaded nanoparticles | Enhances drug efficacy in CRC | Ref. 54 |
| PORCN inhibitor | Suppresses growth of RNF43‐mutant cell‐derived PDX | Ref. 104 |
| Statins | Displays potent roles for APC‐mutated CRC | Ref. 105 |
| S63845 | Overcomes intrinsic and acquired regorafenib resistance in CRC | Ref. 106 |
Abbreviations: CRC, colorectal cancer; G6PD, glucose‐6‐phosphate dehydrogenase; PDX, patient‐derived xenograft; TIMP‐2, tissue inhibitor of matrix metalloproteinase‐2.
Various chemotherapy administration methods have been applied for CRC treatment. The first‐line chemotherapeutic drugs in the treatment of CRC contain 5‐fluorouracil (5‐Fu), oxaliplatin, and irinotecan. 82 CRC often develops chemoresistance with progression, which leads to treatment failure. 5 , 83 The discovery of new drugs to ameliorate CRC chemoresistance is helpful to improve the prognosis of CRC patients. It has been found that interferon‐α could function in enhancing the antitumor activity of 5‐Fu in CRC PDX. 84 Pharmacological targeting protein tyrosine kinase 6 attenuates the chemoresistance of CRC cells by inhibiting Janus kinase 2/signal transducer and activator of transcription 3 pathway in the PDX model. 85 Inhibition of glucose‐6‐phosphate dehydrogenase increases oxaliplatin‐based chemotherapy in the CRC PDX models. 86 Dasatinib, an Src inhibitor, sensitizes liver metastatic CRC to oxaliplatin in CRC with high levels of phospho‐Src using PDX. 87 Further study also shows that dasatinib exhibits modest antitumor effect only when combined with chemotherapy in CRC PDX. 88 It is reported that TIMP‐2 (tissue inhibitor of matrix metalloproteinase‐2) is upregulated in 5‐Fu‐resistant CRC PDX, and inhibition of TIMP‐2 using an anti‐TIMP‐2 antibody or ERK/MAPK (extracellular regulated protein kinase/mitogen‐activated protein kinase) inhibition by U0126 suppresses TIMP‐2‐mediated 5‐Fu‐resistance. 89 The ATM (ataxia telangiectasia‐mutated gene) inhibitor, AZ31, exhibits antitumor activity in CRC PDX resistant to irinotecan monotherapy. 90 Alisertib, an inhibitor of aurora kinases, shows antitumor activity in combination with cetuximab or irinotecan. 91
Recently, anti‐EGFR (epidermal growth factor receptor) monoclonal antibody has been used as the first‐line targeted therapy for CRC. KRAS and NRAS mutations, occurring in 45% of CRC, cause primary resistance to anti‐EGFR therapy. 92 , 93 Thus, it is important to screen potential drugs for KRAS or NRAS mutant CRC using the PDX platform. GC1118, an anti‐EGFR antibody, exhibits promising therapeutic efficacy against KRAS mutation‐driven PDX. 94 TAK‐960, the inhibitor of Polo‐like kinase 1, is an active antiproliferative agent against PDX models carrying KRAS mutation. 95 Research from a novel individualized CRC PDX system revealed that the combination of the inhibitors toward EGFR and MEK (mitogen‐activated protein kinase 1) or CDK4/6 (cyclin‐dependent kinase 4/6) pathway displays a synergistic inhibitory effect against CRC using high‐throughput drug screening. 96 CRISPR screenings uncover a novel combination treatment targeting BCL‐XL and WNT signaling for KRAS/BRAF‐mutated CRC PDX. 97 The combination of bortezomib and everolimus (RAD001) is efficacious against mutant KRAS CRC PDX. 98 RGX‐202, an oral small‐molecule SLC6A8 (solute carrier family 6, member 8) transporter inhibitor, suppresses CRC growth in KRAS mutant CRC PDX. 99 Metformin selectively inhibits metastatic CRC with the KRAS mutation in the PDX model, and retrospective clinical studies further supported the role of metformin. 100
Other inhibitors also showed the potent antitumor activities using CRC PDX. c‐Met inhibitor in combination with anti‐VEGF synergistically reduces microvessel density, suppresses proliferation, and increases apoptosis in c‐Met‐overexpressing CRC PDX. 101 ENMD‐2076, a novel orally active small‐molecule multikinase inhibitor, exhibits a promising antitumor activity in CRC PDX. 102 A nanoparticle‐based agent targeting LRP‐1 (low‐density lipoprotein‐related protein 1) can be used to promote the efficacy of neo‐adjuvant radiotherapy using CRC PDX. 103 SN38‐loaded nanoparticles are capable of enhancing drug efficacy in CRC. 54 PORCN inhibitor significantly suppresses RNF43‐mutant cell‐derived PDX CRC development. 104 Statins, the HMG‐CoA reductase inhibitors, play potent roles in APC‐mutated CRC. 105 Mcl‐1 inhibitors overcome intrinsic and acquired regorafenib resistance in CRC. 106
Advancements in high‐throughput analysis and bioinformatics contribute to cancer research, 107 , 108 enabling PDX to become a powerful preclinical tool for large‐scale drug discovery. 109 Recent studies have suggested that PDXs faithfully recapitulate human tumor biology and predict drug response by directly comparing drug responses in patients and their corresponding xenografts in high‐throughput analysis. 49 , 110 , 111 , 112 High‐throughput drug screening in CRC PDX suggested that the sequential administration of XPO1 (exportin 1) and ATR (ATM and Rad3 related) inhibitors upregulates the therapeutic outcome in TP53‐mutated CRC. 113 Bortezomib, the NF‐κB inhibitor, was selected to render BETi‐resistant cells more sensitive to BETi. 114 PDX is also a promising tool used in uncovering novel immunotherapy methods such as chimeric antigen receptor T cells, which are genetically modified T cells recognizing cancer cells. 115 , 116
Overall, these studies identify PDX as a useful model for conducting in vivo testing of more precise therapy for CRC. Most of these studies employed PDX as a preclinical model for drug screening in the laboratory. Clinical trials to analyze the utility of PDX models are ongoing in human cancers, including CRC (Table 4). These studies will uncover the potential effect of leveraging annotated PDX models matched with the patients in a clinical trial, and this approach will accelerate the development of new drugs and improvement of CRC therapeutics.
TABLE 4.
Clinical trials using PDX model
| Number | Interventions | Conditions | Status |
|---|---|---|---|
| NCT03358628 | In vivo drug testing | Osteosarcoma | Not yet recruiting |
| NCT04602702 | Drug paneling for personalized medicine | Kidney neoplasm, renal cell carcinoma, metastatic solid tumor | Recruiting |
| NCT02247037 | Chemotherapy | Triple negative breast cancer | Completed |
| NCT02720796 | Drug sensitivity testing | Sarcoma | Terminated |
| NCT02910895 | Tumor biopsy | Soft tissue sarcoma | Recruiting |
| NCT02752893 | Biospecimen collection | Breast cancer | Completed |
| NCT04410302 | Biospecimen collection | Bladder carcinoma, gastric carcinoma, liver and intrahepatic bile duct carcinoma | Recruiting |
| NCT02572778 | Local biopsy in the tumor | Squamous cell carcinoma of the head and neck | Recruiting |
| NCT03786848 | MiniPDX group | Prostate cancer | Unknown |
| NCT04703244 | Chemotherapy or endocrine therapy for breast cancer | Breast cancer, residual | Recruiting |
| NCT02732860 | In vivo drug testing in pPDX | Colorectal neoplasms, colorectal cancer, breast cancer, breast neoplasms, ovarian cancer, ovarian neoplasm | Recruiting |
| NCT03219047 | Ibrutinib | Recurrent mantle cell lymphoma, refractory mantle cell lymphoma | Unknown |
| NCT03134456 | Pembrolizumab injection | Metastatic non‐small cell lung carcinoma | Recruiting |
| NCT03551457 | Diagnostic test: biopsy | Urogenital neoplasms | Completed |
| NCT02752932 | Drug testing on PDX | Head and neck squamous cell carcinoma | Completed |
| NCT03336931 | Molecular profiling and drug testing | Childhood cancer, childhood solid tumor, childhood brain tumor, childhood leukemia, refractory cancer, relapsed cancer | Recruiting |
| NCT01750164 | Biospecimen collection | Metastatic breast cancer | Terminated |
| NCT05092009 | Tissue and blood | Lung cancer | Recruiting |
Abbreviation: PDX, patient‐derived xenograft.
4.2. Biomarker discovery
Targeted therapy is helpful to improve the survival of many patients with malignant CRC. 117 The PDX model is developed to detect biomarkers that can effectively predict suitable drugs for specific subgroups of CRC patients. 58 , 73 , 118 A cohort of CRC PDX showed that IGF2 (insulin growth factor 2) may be used as a biomarker of reduced tumor sensitivity to anti‐EGFR therapy and a determinant of response to combined IGF2 and EGFR targeting in CRC patients. 119 Drug screening in a cohort of 49 CRC PDX models revealed that the expression of EGFR ligands was associated with the sensitivity of CRC toward cetuximab. 120 Moreover, the downregulation in classical EGFR‐mediated signaling and the upregulation in antiapoptotic signaling are associated with acquired cetuximab resistance. 120 A recent study reported the dynamic alterations of biomarkers in KRAS G13D mutant CRC PDX model treated with cetuximab. 121 The expression of EGFR ligands is associated with the sensitivity of CRC to cetuximab, and the gene copy number of some oncogenes (BRAF, EGFR, and KRAS) is correlated positively with cetuximab and erlotinib sensitivity. 120 , 122 The implementing systems and molecular images were established to predict the efficacy of BCL‐2 inhibition in CRC based on a PDX cohort. 123 PP2AC expression accurately indicates therapeutic response to p38 inhibitor in CRC PDX. 124 APC and TP53 mutation as convenient biomarkers predicts the sensitivity to EGFR‐targeted therapies. 125 As the key markers are continuously screened out to evaluate the related treatment in CRC PDX models, more targeted drugs are also used more precisely.
4.3. Basic research
PDX models are frequently used in basic research to study the related mechanism in CRC development and progression. Based on PDX metastasis models, researchers found that N6‐methyladenosine modification of circNSUN2 promotes colorectal liver metastasis. 126 METTL3 (methyltransferase like 3) promotes tumor progression via an m6A‐IGF2BP2‐dependent mechanism in CRC. 127 LncRNA LINRIS stabilizes IGF2BP2 (IGF2 binding protein 2) and enhances aerobic glycolysis in CRC. 128 PCK1 (phosphoenolpyruvate carboxykinase 1) and DHODH (dihydroorotate dehydrogenase) participate in CRC liver metastatic colonization by promoting nucleotide synthesis. 129 Inhibition of long noncoding RNA SNHG29‐mediated YAP activation promotes antitumor immunity in CRC. 130 METTL3‐mediated m6A modification of LBX2‐AS1 regulates CRC proliferation, migration, and invasion acting as a ceRNA to sponge miR‐422a. 131 These studies confirmed the roles of RNA‐associated modulation in CRC development and progression using the PDX model. Vesicle transporter GOLT1B (golgi transport 1 homolog B) induces the cell membrane localization of DVL2 (disheveled segment polarity protein 2) and PD‐L2 to promote CRC metastasis. 132 Eukaryotic initiation factor 4A2 promotes cancer cell metastasis and oxaliplatin resistance in CRC. 133 Hypoxia‐induced exosomal miR‐135a‐5p is involved in the development, clinical severity, and prognosis of CRC liver metastases through the pre‐metastatic niche. 134 LncRNA LVBU promotes CRC progression by regulating urea cycle/polyamine synthesis axis. 135 These papers studied the mechanism of CRC metastasis using the PDX model. Overall, PDX models are becoming more common in CRC basic research.
5. ADVANTAGES OF CRC PDX MODEL
Currently, many studies have reported that the CRC PDX model maintains the genetic consistency with the primary tumors. 136 , 137 , 138 , 139 , 140 The comprehensive genetic characterization of PDXs in CRC uncovered that the frequency of key genetic mutations in PDXs is similar to that reported in primary tumors. The common genetic mutations and their frequencies, such as APC (75.9%), TP53 (70.9%), KRAS (55.7%), NRAS (5.1%), BRAF (15.2%), PIK3CA (26.6%), PIK3R1 (6.3%), and CTNNB1 (3.8%), are consistent with the data reported in human CRC tumors in a cohort of 79 CRC PDX models by whole‐exome and RNA sequencing. 136 This consistency is important for drug screening, biomarker identification, and basic research. To some extent, the PDX model can also simulate the interactions between cancer cells and TME, which plays pivotal roles in tumor progression. 73 Recent studies observed one of the molecular subtypes (CMS4), which is characterized by the activation of pathways related to EMT and stemness, such as TGF‐β and integrin, and is mostly regulated by prominent stromal cell infiltration of adjacent cancer tissue, particularly CAFs in CRC. 137 , 141 , 142 The high expression of TME signatures plays an important role in inducing resistance of cancer cells to chemotherapies and targeted agents in CMS4 CRC. 141 , 142 , 143 Moreover, the TGF‐β signaling inhibitors exhibit the ability to block the cross‐talk between cancer cells and the microenvironment and reduce the number of metastases in CRC PDX models, indicating a dependency on TGF‐β signaling in stromal cells during metastasis in CRC. 143 , 144 The retrospective analysis of a randomized clinical study also suggested that the CMS4 subtype exhibits a lack of benefit from adjuvant oxaliplatin‐based chemotherapy in CRC patients. 145 Thus, PDX models can be used to evaluate the strategies targeting TME.
Recent evidences have demonstrated that PDX models exhibit good abilities to predict the outcomes of both traditional and novel antitumor therapeutics, revealing that PDX could be used in “co‐clinical trials” for screening drugs. 118 , 146 After the PDX models were given the same treatment as the primary patients, they accurately replicated the patients' clinical outcomes, even as the patients underwent several additional cycles of therapy over time. 147 Thus, if the investigations in in vivo tests and in clinical trials are performed in parallel, we can identify therapeutic targets more rapidly, especially for some rare types of cancers. Furthermore, the methods of “co‐clinical trials” can be used to seek more appropriate strategies for cancer patients based on the drug sensitivity of the PDX model. In this situation, PDX models can be applied as an “avatar model”. 148 Because gene signature alone is insufficient to uncover therapeutic options for the majority of patients with advanced disease, the generation of the PDX platform contributes to the discovery of novel therapeutic approaches that potentially provide personalized therapeutic options for individual patients. 149 , 150 These studies identified the PDX system as an important tool for precise medicine because it can highly reproduce the outcomes of antitumor agents in patients.
6. LIMITATIONS OF CRC PDX MODEL
One problem in the development of the PDX model is engraftment failure. The rate of successful PDX engraftment is relatively high for CRC, ranging from 56% to 87.5% according to published studies, 58 , 151 , 152 , 153 compared to other cancers, such as epithelial ovarian cancer (48.8%) and breast cancer (27%). 154 , 155 The success rate of CRC PDX engraftment is higher in surgical specimens (36/50, 72%) versus in biopsy specimens (14/40, 35%). 151 The selection of mouse model is also important, with more severely immunosuppressed strains such as NOD/SCID or NOD/SCID/IL2gnull (NSG) being easier for PDX construction, due to their lower immune rejection to engraftment. 60 , 76
Although the CRC PDX platform is highly informative, some issues need to be further addressed. It is likely that PDX cannot fully represent the heterogeneity of the primary tumor. It has been shown that human tumors consist of multiple subclones carrying distinct genetic and epigenetic alterations, especially for metastatic CRCs. 156 , 157 , 158 It is impossible to cover this intra‐tumoral heterogeneity because it is difficult to avoid the sampling bias. Moreover, the human TME is rapidly replaced in a time‐ and size‐dependent manner by murine stromal cells, including CAFs, inflammatory cells, extracellular matrix, and blood/lymphatic vessels over time. 159 , 160 The strategy for overcoming this problem is to develop the humanized PDX model. Briefly, the human peripheral blood cells, hematopoietic stem cells, or human tumor‐infiltrating lymphocytes are implanted into immune‐deficient mice after ablating the endogenous immune system for humanization. 161 , 162 In recent years, tumor immunotherapy using different approaches, including specific antibodies, immune checkpoint blockades, cancer vaccines, and adoptive cell therapies, has been widely recognized as an effective and promising method with fewer side effects. 163 , 164 , 165 The PDX model lacks a complete immune system and is not suitable for immunotherapy research. Humanized mice are developed to address this issue. However, these models simulate only some aspects of the human immune system, failing to reproduce a functional and complete human immune system.
Orthotopic models provide a more suitable platform to study the biology of metastasis and response to therapy in CRC. 11 , 166 Orthotopic implantation into the colorectal wall of the host requires considerable technical skill. Thus, the difficulty of orthotopic transplantation also limits the capacity and reproducibility. The establishment and management of large PDX cohorts need vast labor and resource. It generally takes 1–4 months to construct a PDX model and 4–8 months to expand a sufficient number of mice to perform an interpretable preclinical study. 167 Typically, it is impossible for patients with advanced cancer to wait 4–8 months for clinical decision making based on PDX model drug tests.
Because PDX trials are technically cumbersome, time consuming, and expensive, in vitro cultures of patient‐derived cancer cells show the potential to be more easily expanded for genetic manipulations and high‐throughput screenings, which are called patient‐derived organoids (PDOs) (Figure 4). 11 , 168 Recently, it has been optimized how to establish PDOs. 63 , 169 , 170 , 171 This approach allows self‐organizing three‐dimensional structures that reconstitute multiple functional structures of the primary tumors. 170 , 172 It is reported that primary or metastatic CRC PDOs can be directly established from patient specimens (biopsy or surgical resection) or PDX explants. 173 , 174 , 175 The success rates of CRC PDOs range between 60% and 90% according to the reports, and CRC PDOs inherit many features of the original tumor tissues, including histopathologic, genetic, and transcriptomic profiles. 176 , 177 , 178 , 179 , 180 Thus, PDOs might be an alternative option to the PDX model. With the development of novel preclinical models such as PDX and PDO, CRC treatment will eventually move to the era of individualized treatment.
FIGURE 4.

The establishment of PDOs (patient‐derived organoids). The tumor tissues derived from CRC (colorectal cancer) patients are cultured under three‐dimensional conditions. The organoids are then obtained, which have many features similar to the original tumor tissues in patients. Thus, these organoids can be used to mimic the tumors in patients
7. PROSPECTS
CRC is a multistep disease caused by the combination of various genetic and epigenetic alterations and environmental risk factors. The development of new therapeutic methods for CRC remains an ongoing challenge because of the inter‐ and intra‐tumoral phenotypic heterogeneity characterized by distinct molecular traits and responses to therapy. 9 However, phenotypic heterogeneity is not reflected in traditional CRC models such as tumor cell lines and GEMMs. The PDX system is a promising tool to study the development and progression of CRC and uncover the latent mechanisms. Ideally, the preclinical model in oncology should reach some criteria to help clinical decisions: availability in number, variations in genetic background, maintenance of the characteristics of human tumors, and consistency in therapy response similar to patients. 73 , 118 The large, high‐quality PDX biobanks are helpful for supporting further studies (Figure 5). Currently, some international groups are engaged in establishing the PDX biobank. These groups include EurOPDX, the NCI Repository of Patient‐Derived Models, the Innovative MODels Initiative (IMODI) Consortium, the Pediatric Preclinical Testing Consortium, the Children's Oncology Group Cell Culture and Xenograft Repository, the Public Repository of Xenografts, and the Jackson Laboratory PDX Resource. 64 , 181 These great efforts will promote the application of PDX models in drug discovery and basic research in the context of population‐scale studies to improve the prognosis of cancer patients. 182 Although a large number of Chinese groups have made efforts to develop the PDX model for single‐center research, 126 , 130 , 183 , 184 , 185 integrative cohorts containing multicenter data should be established and maintained in the future.
FIGURE 5.

The establishment of PDX (patient‐derived xenograft) biobank for precision medicine. Specimens from CRC patients enrolled in the study are submitted to PDX biobank. Researchers can take advantage of the biobanks to screen drugs, uncover biomarkers, and study the basic mechanisms. The latent drugs will submit to animal tests using PDX in biobanks. The effective drugs and biomarkers will used to improve the therapeutic effects.
Standardizing protocols for constructing and biobanking PDX models are critical to maximize the impact of studies using these publically available PDX resources. Currently, the fact that underlying mechanisms about the occurrence and progression of CRC are not fully elucidated causes the difficulty in the standardization of the CRC PDX model system. In future studies, with the development of high‐throughput sequencing and other technologies, 186 it will be much easier to construct the CRC PDX model for simulating the heterogeneity of human CRC tumors. In addition, it is important to study how to reduce the time to establish the CRC PDX model. Improved transplantation technologies, such as ultrasound‐guided fine needle aspiration, may be helpful. Clinically, ultrasound‐guided fine needle aspiration technique is commonly employed for tissue acquisition. 187 The ultrasound monitor is applied as a reference to orthotopically implant CRC cells into mice. Orthotopic implantation contributes to increase in the success rate of the PDX model. Because of its minimally invasive feature, this method decreases the risk of complications in orthotopic transplantation. Compared with the traditional orthotopic transplantation model, the occurrence of inflammation is reduced and the animal's healing time is reduced, which significantly upregulate the survival rate of mice. Currently, attempts have been made to establish the PDX model of some types of cancers using ultrasound monitor, such as lung cancer, pancreatic cancer, and melanoma. 188 , 189
In conclusion, the PDX model has emerged as a promising approach in the discovery of oncologic drugs and basic research in CRC. The PDX model is widely used in various CRC research studies. However, this model has certain limitations, including the use of animals, limited engraftment efficiency, and high costs. These issues need to be further addressed. Now, the PDO model is established and can be used to overcome these limitations to some extent.
AUTHOR CONTRIBUTIONS
Xiaofeng Liu and Zechang Xin prepared the manuscript. Kun Wang reviewed the manuscript.
CONFLICT OF INTEREST
The authors have no conflicts of interest. Xiaofeng Liu is an editorial board member of AMEM and a coauthor of this article. To minimize bias, he was excluded from all editorial decision making related to the acceptance of this article for publication.
ETHICS APPROVAL
All authors read and approved the final manuscript.
ACKNOWLEDGMENT
The work was supported by the National Natural Science Foundation of China Grant (81802305 and 31971192).
Liu X, Xin Z, Wang K. Patient‐derived xenograft model in colorectal cancer basic and translational research. Anim Models Exp Med. 2023;6:26‐40. doi: 10.1002/ame2.12299
Funding information
National Natural Science Foundation of China Grant (81802305 and 31971192).
Contributor Information
Xiaofeng Liu, Email: liuxiaofeng100@bjmu.edu.cn.
Kun Wang, Email: wang-kun@vip.sina.com.
REFERENCES
- 1. Dekker E, Tanis PJ, Vleugels JLA, Kasi PM, Wallace MB. Colorectal cancer. Lancet. 2019;394:1467‐1480. [DOI] [PubMed] [Google Scholar]
- 2. Xu D, Liu XF, Yan XL, Wang K, Xing BC. Survival prediction in patients with resectable colorectal liver metastases: clinical risk scores and tumor response to chemotherapy. Oncol Lett. 2017;14:8051‐8059. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Liu W, Wang K, Han Y, Liang JY, Li YH, Xing BC. Nomogram predicted disease free survival for colorectal liver metastasis patients with preoperative chemotherapy followed by hepatic resection. Eur J Surg Oncol. 2019;45:2070‐2077. [DOI] [PubMed] [Google Scholar]
- 4. Wagner J, Wickman E, DeRenzo C, Gottschalk S. CAR T cell therapy for solid tumors: bright future or dark reality? Mol Ther. 2020;28:2320‐2339. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Liu X, Su K, Sun X, et al. Sec62 promotes stemness and chemoresistance of human colorectal cancer through activating Wnt/beta‐catenin pathway. J Exp Clin Cancer Res. 2021;40:132. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Tang Q, Chen J, Di Z, et al. TM4SF1 promotes EMT and cancer stemness via the Wnt/beta‐catenin/SOX2 pathway in colorectal cancer. J Exp Clin Cancer Res. 2020;39:232. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Liu Z, Liu X, Li Y, et al. miR‐6716‐5p promotes metastasis of colorectal cancer through downregulating NAT10 expression. Cancer Manag Res. 2019;11:5317‐5332. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Mori G, Pasca MR. Gut microbial signatures in sporadic and hereditary colorectal cancer. Int J Mol Sci. 2021;22:1312. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. El Bali M, Bakkach J, Bennani Mechita M. Colorectal cancer: from genetic landscape to targeted therapy. J Oncol. 2021;2021:9918116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Zhang JY, Ren PW, Xu D, et al. Human UTP14a promotes colorectal cancer progression by forming a positive regulation loop with c‐Myc. Cancer Lett. 2019;440:106‐115. [DOI] [PubMed] [Google Scholar]
- 11. Rizzo G, Bertotti A, Leto SM, Vetrano S. Patient‐derived tumor models: a more suitable tool for pre‐clinical studies in colorectal cancer. J Exp Clin Cancer Res. 2021;40:178. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Li Y, Wang L, Liu X, Zhang C, Du X, Xing B. NIR promotes progression of colorectal cancer through regulating RB. Biochim Biophys Acta Mol Cell Res. 2021;1868:118856. [DOI] [PubMed] [Google Scholar]
- 13. Liu X, Tan Y, Zhang C, et al. NAT10 regulates p53 activation through acetylating p53 at K120 and ubiquitinating Mdm2. EMBO Rep. 2016;17:349‐366. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Greaves M. Evolutionary determinants of cancer. Cancer Discov. 2015;5:806‐820. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Jung G, Hernandez‐Illan E, Moreira L, Balaguer F, Goel A. Epigenetics of colorectal cancer: biomarker and therapeutic potential. Nat Rev Gastroenterol Hepatol. 2020;17:111‐130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Bazzichetto C, Conciatori F, Falcone I, Cognetti F, Milella M, Ciuffreda L. Advances in tumor‐stroma interactions: emerging role of cytokine network in colorectal and pancreatic cancer. J Oncol. 2019;2019:5373580. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Huang C, Ou R, Chen X, et al. Tumor cell‐derived SPON2 promotes M2‐polarized tumor‐associated macrophage infiltration and cancer progression by activating PYK2 in CRC. J Exp Clin Cancer Res. 2021;40:304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Tan Y, Zheng J, Liu X, et al. Loss of nucleolar localization of NAT10 promotes cell migration and invasion in hepatocellular carcinoma. Biochem Biophys Res Commun. 2018;499:1032‐1038. [DOI] [PubMed] [Google Scholar]
- 19. Neophytou CM, Panagi M, Stylianopoulos T, Papageorgis P. The role of tumor microenvironment in cancer metastasis: molecular mechanisms and therapeutic opportunities. Cancers (Basel). 2021;13:2235. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Wan PK, Ryan AJ, Seymour LW. Beyond cancer cells: targeting the tumor microenvironment with gene therapy and armed oncolytic virus. Mol Ther. 2021;29:1668‐1682. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Fang K, Zhan Y, Zhu R, et al. Bufalin suppresses tumour microenvironment‐mediated angiogenesis by inhibiting the STAT3 signalling pathway. J Transl Med. 2021;19:383. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Gallo G, Vescio G, De Paola G, Sammarco G. Therapeutic targets and tumor microenvironment in colorectal cancer. J Clin Med. 2021;10:2295. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Hait WN. Anticancer drug development: the grand challenges. Nat Rev Drug Discov. 2010;9:253‐254. [DOI] [PubMed] [Google Scholar]
- 24. Bellio H, Fumet JD, Ghiringhelli F. Targeting BRAF and RAS in colorectal cancer. Cancers (Basel). 2021;13:2201. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Rech AJ, Mick R, Martin S, et al. CD25 blockade depletes and selectively reprograms regulatory T cells in concert with immunotherapy in cancer patients. Sci Transl Med. 2012;4:134ra162. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Guo J, Yu Z, Sun D, Zou Y, Liu Y, Huang L. Two nanoformulations induce reactive oxygen species and immunogenetic cell death for synergistic chemo‐immunotherapy eradicating colorectal cancer and hepatocellular carcinoma. Mol Cancer. 2021;20:10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Li C, Lau HC, Zhang X, Yu J. Mouse models for application in colorectal cancer: understanding the pathogenesis and relevance to the human condition. Biomedicine. 2022;10:1710. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Chen S, Zhang L, Li M, et al. Fusobacterium nucleatum reduces METTL3‐mediated m(6)a modification and contributes to colorectal cancer metastasis. Nat Commun. 2022;13:1248. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Xin B, Yang M, Wu P, et al. Enhancing the therapeutic efficacy of programmed death ligand 1 antibody for metastasized liver cancer by overcoming hepatic immunotolerance in mice. Hepatology. 2022;76:630‐645. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Wagner S, Beger NT, Matschos S, et al. Tumour‐derived cell lines and their potential for therapy prediction in patients with metastatic colorectal cancer. Cancers (Basel). 2021;13:4717. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Betzler AM, Nanduri LK, Hissa B, et al. Differential effects of Trp53 alterations in murine colorectal cancer. Cancers (Basel). 2021;13:808. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Shao L, Guo YP, Wang L, et al. Effects of ginsenoside compound K on colitis‐associated colorectal cancer and gut microbiota profiles in mice. Ann Transl Med. 2022;10:408. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Moser AR, Pitot HC, Dove WF. A dominant mutation that predisposes to multiple intestinal neoplasia in the mouse. Science. 1990;247:322‐324. [DOI] [PubMed] [Google Scholar]
- 34. Oliveira RC, Abrantes AM, Tralhao JG, Botelho MF. The role of mouse models in colorectal cancer research‐the need and the importance of the orthotopic models. Animal Model Exp Med. 2020;3:1‐8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Parang B, Barrett CW, Williams CS. AOM/DSS model of colitis‐associated cancer. Methods Mol Biol. 2016;1422:297‐307. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Zhang Y, Pu W, Bousquenaud M, et al. Emodin inhibits inflammation, carcinogenesis, and cancer progression in the AOM/DSS model of colitis‐associated intestinal tumorigenesis. Front Oncol. 2020;10:564674. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. De Robertis M, Massi E, Poeta ML, et al. The AOM/DSS murine model for the study of colon carcinogenesis: from pathways to diagnosis and therapy studies. J Carcinog. 2011;10:9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Burtin F, Mullins CS, Linnebacher M. Mouse models of colorectal cancer: past, present and future perspectives. World J Gastroenterol. 2020;26:1394‐1426. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Shen X, Jain A, Aladelokun O, et al. Asparagine, colorectal cancer, and the role of sex, genes, microbes, and diet: a narrative review. Front Mol Biosci. 2022;9:958666. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Dos Santos W, Dos Reis MB, Porto J, et al. Somatic targeted mutation profiling of colorectal cancer precursor lesions. BMC Med Genomics. 2022;15:143. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Yang Y, Ren P, Liu X, et al. PPP1R26 drives hepatocellular carcinoma progression by controlling glycolysis and epithelial‐mesenchymal transition. J Exp Clin Cancer Res. 2022;41:101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Zheng J, Tan Y, Liu X, et al. NAT10 regulates mitotic cell fate by acetylating Eg5 to control bipolar spindle assembly and chromosome segregation. Cell Death Differ. 2022;29:846‐860. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Li Q, Liu X, Jin K, et al. NAT10 is upregulated in hepatocellular carcinoma and enhances mutant p53 activity. BMC Cancer. 2017;17:605. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Cai S, Liu X, Zhang C, Xing B, Du X. Autoacetylation of NAT10 is critical for its function in rRNA transcription activation. Biochem Biophys Res Commun. 2017;483:624‐629. [DOI] [PubMed] [Google Scholar]
- 45. Liu XF, Cai SY, Zhang CF, et al. Deacetylation of NAT10 by Sirt1 promotes the transition from rRNA biogenesis to autophagy upon energy stress. Nucleic Acids Res. 2018;46:9601‐9616. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Chen M, Zhang C, Liu W, Du X, Liu X, Xing B. Long noncoding RNA LINC01234 promotes hepatocellular carcinoma progression through orchestrating aspartate metabolic reprogramming. Mol Ther. 2022;30:2354‐2369. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Medico E, Russo M, Picco G, et al. The molecular landscape of colorectal cancer cell lines unveils clinically actionable kinase targets. Nat Commun. 2015;6:7002. [DOI] [PubMed] [Google Scholar]
- 48. Ben‐David U, Siranosian B, Ha G, et al. Genetic and transcriptional evolution alters cancer cell line drug response. Nature. 2018;560:325‐330. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Hidalgo M, Amant F, Biankin AV, et al. Patient‐derived xenograft models: an emerging platform for translational cancer research. Cancer Discov. 2014;4:998‐1013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Shi L, Wang Y, Wang Q, et al. Transforming a toxic drug into an efficacious nanomedicine using a lipoprodrug strategy for the treatment of patient‐derived melanoma xenografts. J Control Release. 2020;324:289‐302. [DOI] [PubMed] [Google Scholar]
- 51. Wang H, Zhou L, Xie K, et al. Polylactide‐tethered prodrugs in polymeric nanoparticles as reliable nanomedicines for the efficient eradication of patient‐derived hepatocellular carcinoma. Theranostics. 2018;8:3949‐3963. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Ren L, Xu P, Yao J, et al. Targeting the mitochondria with pseudo‐stealthy Nanotaxanes to impair mitochondrial biogenesis for effective cancer treatment. ACS Nano. 2022;16:10242‐10259. [DOI] [PubMed] [Google Scholar]
- 53. Okada S, Vaeteewoottacharn K, Kariya R. Application of highly immunocompromised mice for the establishment of patient‐derived xenograft (PDX) models. Cell. 2019;8:889. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Wang Y, Xie H, Ying K, et al. Tuning the efficacy of esterase‐activatable prodrug nanoparticles for the treatment of colorectal malignancies. Biomaterials. 2021;270:120705. [DOI] [PubMed] [Google Scholar]
- 55. Huang L, Chen X, Bian Q, et al. Photosensitizer‐stabilized self‐assembling nanoparticles potentiate chemo/photodynamic efficacy of patient‐derived melanoma. J Control Release. 2020;328:325‐338. [DOI] [PubMed] [Google Scholar]
- 56. Fu XY, Besterman JM, Monosov A, Hoffman RM. Models of human metastatic colon cancer in nude mice orthotopically constructed by using histologically intact patient specimens. Proc Natl Acad Sci USA. 1991;88:9345‐9349. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Burgenske DM, Monsma DJ, MacKeigan JP. Patient‐derived xenograft models of colorectal cancer: procedures for engraftment and propagation. Methods Mol Biol. 2018;1765:307‐314. [DOI] [PubMed] [Google Scholar]
- 58. Puig I, Chicote I, Tenbaum SP, et al. A personalized preclinical model to evaluate the metastatic potential of patient‐derived colon cancer initiating cells. Clin Cancer Res. 2013;19:6787‐6801. [DOI] [PubMed] [Google Scholar]
- 59. AlMusawi S, Ahmed M, Nateri AS. Understanding cell‐cell communication and signaling in the colorectal cancer microenvironment. Clin Transl Med. 2021;11:e308. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Brown KM, Xue A, Mittal A, Samra JS, Smith R, Hugh TJ. Patient‐derived xenograft models of colorectal cancer in pre‐clinical research: a systematic review. Oncotarget. 2016;7:66212‐66225. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Bertotti A, Migliardi G, Galimi F, et al. A molecularly annotated platform of patient‐derived xenografts ("xenopatients") identifies HER2 as an effective therapeutic target in cetuximab‐resistant colorectal cancer. Cancer Discov. 2011;1:508‐523. [DOI] [PubMed] [Google Scholar]
- 62. Jung J, Kim J, Lim HK, et al. Establishing a colorectal cancer liver metastasis patient‐derived tumor xenograft model for the evaluation of personalized chemotherapy. Ann Surg Treat Res. 2017;93:173‐180. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Bleijs M, van de Wetering M, Clevers H, Drost J. Xenograft and organoid model systems in cancer research. EMBO J. 2019;38:e101654. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Gao H, Korn JM, Ferretti S, et al. High‐throughput screening using patient‐derived tumor xenografts to predict clinical trial drug response. Nat Med. 2015;21:1318‐1325. [DOI] [PubMed] [Google Scholar]
- 65. Cybulska M, Olesinski T, Goryca K, et al. Challenges in stratifying the molecular variability of patient‐derived colon tumor xenografts. Biomed Res Int. 2018;2018:2954208. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Prasetyanti PR, van Hooff SR, van Herwaarden T, et al. Capturing colorectal cancer inter‐tumor heterogeneity in patient‐derived xenograft (PDX) models. Int J Cancer. 2019;144:366‐371. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Cho YB, Hong HK, Choi YL, et al. Colorectal cancer patient‐derived xenografted tumors maintain characteristic features of the original tumors. J Surg Res. 2014;187:502‐509. [DOI] [PubMed] [Google Scholar]
- 68. Lee WS, Kim HY, Seok JY, et al. Genomic profiling of patient‐derived colon cancer xenograft models. Medicine (Baltimore). 2014;93:e298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Meehan TF, Conte N, Goldstein T, et al. PDX‐MI: minimal information for patient‐derived tumor xenograft models. Cancer Res. 2017;77:e62‐e66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Jung J, Seol HS, Chang S. The generation and application of patient‐derived xenograft model for cancer research. Cancer Res Treat. 2018;50:1‐10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Hoffman RM. Patient‐derived orthotopic xenografts: better mimic of metastasis than subcutaneous xenografts. Nat Rev Cancer. 2015;15:451‐452. [DOI] [PubMed] [Google Scholar]
- 72. Visioli A, Giani F, Trivieri N, et al. Stemness underpinning all steps of human colorectal cancer defines the core of effective therapeutic strategies. EBioMedicine. 2019;44:346‐360. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Inoue A, Deem AK, Kopetz S, Heffernan TP, Draetta GF, Carugo A. Current and future horizons of patient‐derived xenograft models in colorectal cancer translational research. Cancers (Basel). 2019;11:1321. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Nemati F, Sastre‐Garau X, Laurent C, et al. Establishment and characterization of a panel of human uveal melanoma xenografts derived from primary and/or metastatic tumors. Clin Cancer Res. 2010;16:2352‐2362. [DOI] [PubMed] [Google Scholar]
- 75. Day CP, Merlino G, Van Dyke T. Preclinical mouse cancer models: a maze of opportunities and challenges. Cell. 2015;163:39‐53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Shultz LD, Goodwin N, Ishikawa F, Hosur V, Lyons BL, Greiner DL. Human cancer growth and therapy in immunodeficient mouse models. Cold Spring Harb Protoc. 2014;2014:694‐708. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Xiao J, Glasgow E, Agarwal S. Zebrafish xenografts for drug discovery and personalized medicine. Trends Cancer. 2020;6:569‐579. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Wu JQ, Zhai J, Li CY, et al. Patient‐derived xenograft in zebrafish embryos: a new platform for translational research in gastric cancer. J Exp Clin Cancer Res. 2017;36:160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Costa B, Estrada MF, Mendes RV, Fior R. Zebrafish avatars towards personalized medicine‐a comparative review between avatar models. Cell. 2020;9:293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Fichtner I, Slisow W, Gill J, et al. Anticancer drug response and expression of molecular markers in early‐passage xenotransplanted colon carcinomas. Eur J Cancer. 2004;40:298‐307. [DOI] [PubMed] [Google Scholar]
- 81. Nunes M, Vrignaud P, Vacher S, et al. Evaluating patient‐derived colorectal cancer xenografts as preclinical models by comparison with patient clinical data. Cancer Res. 2015;75:1560‐1566. [DOI] [PubMed] [Google Scholar]
- 82. Ciombor KK, Wu C, Goldberg RM. Recent therapeutic advances in the treatment of colorectal cancer. Annu Rev Med. 2015;66:83‐95. [DOI] [PubMed] [Google Scholar]
- 83. Wang H, Yang W, Qin Q, et al. E3 ubiquitin ligase MAGI3 degrades c‐Myc and acts as a predictor for chemotherapy response in colorectal cancer. Mol Cancer. 2022;21:151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Laurent PL, Tevaearai HT, Eliason JF, Givel JC, Odartchenko N. Interferon alpha and 5′‐deoxy‐5‐fluorouridine in colon cancer: effects as single agents and in combination on growth of xenograft tumours. Eur J Cancer. 1994;30a:1859‐1865. [DOI] [PubMed] [Google Scholar]
- 85. Liu C, Pan Z, Chen Q, et al. Pharmacological targeting PTK6 inhibits the JAK2/STAT3 sustained stemness and reverses chemoresistance of colorectal cancer. J Exp Clin Cancer Res. 2021;40:297. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Ju HQ, Lu YX, Wu QN, et al. Disrupting G6PD‐mediated redox homeostasis enhances chemosensitivity in colorectal cancer. Oncogene. 2017;36:6282‐6292. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. Perez M, Lucena‐Cacace A, Marin‐Gomez LM, et al. Dasatinib, a Src inhibitor, sensitizes liver metastatic colorectal carcinoma to oxaliplatin in tumors with high levels of phospho‐Src. Oncotarget. 2016;7:33111‐33124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Scott AJ, Song EK, Bagby S, et al. Evaluation of the efficacy of dasatinib, a Src/Abl inhibitor, in colorectal cancer cell lines and explant mouse model. PLoS One. 2017;12:e0187173. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Zhang G, Luo X, Wang Z, et al. TIMP‐2 regulates 5‐Fu resistance via the ERK/MAPK signaling pathway in colorectal cancer. Aging (Albany NY). 2022;14:297‐315. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Greene J, Nguyen A, Bagby SM, et al. The novel ATM inhibitor (AZ31) enhances antitumor activity in patient derived xenografts that are resistant to irinotecan monotherapy. Oncotarget. 2017;8:110904‐110913. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Pitts TM, Bradshaw‐Pierce EL, Bagby SM, et al. Antitumor activity of the aurora a selective kinase inhibitor, alisertib, against preclinical models of colorectal cancer. Oncotarget. 2016;7:50290‐50301. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Janssen JBE, Medema JP, Gootjes EC, Tauriello DVF, Verheul HMW. Mutant RAS and the tumor microenvironment as dual therapeutic targets for advanced colorectal cancer. Cancer Treat Rev. 2022;109:102433. [DOI] [PubMed] [Google Scholar]
- 93. Lakatos G, Kohne CH, Bodoky G. Current therapy of advanced colorectal cancer according to RAS/RAF mutational status. Cancer Metastasis Rev. 2020;39:1143‐1157. [DOI] [PubMed] [Google Scholar]
- 94. Lee HW, Son E, Lee K, et al. Promising therapeutic efficacy of GC1118, an anti‐EGFR antibody, against KRAS mutation‐driven colorectal cancer patient‐derived xenografts. Int J Mol Sci. 2019;20:5894. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Klauck PJ, Bagby SM, Capasso A, et al. Antitumor activity of the polo‐like kinase inhibitor, TAK‐960, against preclinical models of colorectal cancer. BMC Cancer. 2018;18:136. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. Wang Y, Liao H, Zheng T, et al. Conditionally reprogrammed colorectal cancer cells combined with mouse avatars identify synergy between EGFR and MEK or CDK4/6 inhibitors. Am J Cancer Res. 2020;10:249‐262. [PMC free article] [PubMed] [Google Scholar]
- 97. Jung HR, Oh Y, Na D, et al. CRISPR screens identify a novel combination treatment targeting BCL‐XL and WNT signaling for KRAS/BRAF‐mutated colorectal cancers. Oncogene. 2021;40:3287‐3302. [DOI] [PubMed] [Google Scholar]
- 98. Ruan H, Leibowitz BJ, Peng Y, et al. Targeting Myc‐driven stress vulnerability in mutant KRAS colorectal cancer. Mol Biomed. 2022;3:10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. Kurth I, Yamaguchi N, Andreu‐Agullo C, et al. Therapeutic targeting of SLC6A8 creatine transporter suppresses colon cancer progression and modulates human creatine levels. Sci Adv. 2021;7:eabi7511. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Xie J, Xia L, Xiang W, et al. Metformin selectively inhibits metastatic colorectal cancer with the KRAS mutation by intracellular accumulation through silencing MATE1. Proc Natl Acad Sci USA. 2020;117:13012‐13022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. Chen X, Guan Z, Lu J, et al. Synergistic antitumor effects of cMet inhibitor in combination with anti‐VEGF in colorectal cancer patient‐derived xenograft models. J Cancer. 2018;9:1207‐1217. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102. Capasso A, Pitts TM, Klauck PJ, et al. Dual compartmental targeting of cell cycle and angiogenic kinases in colorectal cancer models. Anticancer Drugs. 2018;29:827‐838. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Lee KJ, Ko EJ, Park YY, et al. A novel nanoparticle‐based theranostic agent targeting LRP‐1 enhances the efficacy of neoadjuvant radiotherapy in colorectal cancer. Biomaterials. 2020;255:120151. [DOI] [PubMed] [Google Scholar]
- 104. Yamamoto D, Oshima H, Wang D, et al. Characterization of RNF43 frameshift mutations that drive Wnt ligand‐ and R‐spondin‐dependent colon cancer. J Pathol. 2022;257:39‐52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105. Shailes H, Tse WY, Freitas MO, Silver A, Martin SA. Statin treatment as a targeted therapy for APC‐mutated colorectal cancer. Front Oncol. 2022;12:880552. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106. Song X, Shen L, Tong J, et al. Mcl‐1 inhibition overcomes intrinsic and acquired regorafenib resistance in colorectal cancer. Theranostics. 2020;10:8098‐8110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107. Liu X, Xu D, Liu Z, et al. THBS1 facilitates colorectal liver metastasis through enhancing epithelial‐mesenchymal transition. Clin Transl Oncol. 2020;22:1730‐1740. [DOI] [PubMed] [Google Scholar]
- 108. Zhang C, Hu C, Su K, et al. The integrative analysis of thrombospondin family genes in Pan‐cancer reveals that THBS2 facilitates gastrointestinal cancer metastasis. J Oncol. 2021;2021:4405491. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109. Cao J, Chan WC, Chow MSS. Use of conditional reprogramming cell, patient derived xenograft and organoid for drug screening for individualized prostate cancer therapy: current and future perspectives (review). Int J Oncol. 2022;60:52. [DOI] [PubMed] [Google Scholar]
- 110. Tentler JJ, Tan AC, Weekes CD, et al. Patient‐derived tumour xenografts as models for oncology drug development. Nat Rev Clin Oncol. 2012;9:338‐350. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111. Siolas D, Hannon GJ. Patient‐derived tumor xenografts: transforming clinical samples into mouse models. Cancer Res. 2013;73:5315‐5319. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112. Rosfjord E, Lucas J, Li G, Gerber HP. Advances in patient‐derived tumor xenografts: from target identification to predicting clinical response rates in oncology. Biochem Pharmacol. 2014;91:135‐143. [DOI] [PubMed] [Google Scholar]
- 113. Inoue A, Robinson FS, Minelli R, et al. Sequential administration of XPO1 and ATR inhibitors enhances therapeutic response in TP53‐mutated colorectal cancer. Gastroenterology. 2021;161:196‐210. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114. Wu T, Wang G, Chen W, et al. Co‐inhibition of BET proteins and NF‐kappaB as a potential therapy for colorectal cancer through synergistic inhibiting MYC and FOXM1 expressions. Cell Death Dis. 2018;9:315. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115. Feins S, Kong W, Williams EF, Milone MC, Fraietta JA. An introduction to chimeric antigen receptor (CAR) T‐cell immunotherapy for human cancer. Am J Hematol. 2019;94:S3‐S9. [DOI] [PubMed] [Google Scholar]
- 116. Teng R, Zhao J, Zhao Y, et al. Chimeric antigen receptor‐modified T cells repressed solid tumors and their relapse in an established patient‐derived colon carcinoma xenograft model. J Immunother. 2019;42:33‐42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117. Xie YH, Chen YX, Fang JY. Comprehensive review of targeted therapy for colorectal cancer. Signal Transduct Target Ther. 2020;5:22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118. Byrne AT, Alférez DG, Amant F, et al. Interrogating open issues in cancer precision medicine with patient‐derived xenografts. Nat Rev Cancer. 2017;17:254‐268. [DOI] [PubMed] [Google Scholar]
- 119. Zanella ER, Galimi F, Sassi F, et al. IGF2 is an actionable target that identifies a distinct subpopulation of colorectal cancer patients with marginal response to anti‐EGFR therapies. Sci Transl Med. 2015;7:272ra212. [DOI] [PubMed] [Google Scholar]
- 120. Rivera M, Fichtner I, Wulf‐Goldenberg A, et al. Patient‐derived xenograft (PDX) models of colorectal carcinoma (CRC) as a platform for chemosensitivity and biomarker analysis in personalized medicine. Neoplasia. 2021;23:21‐35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121. Zhang H, Yuan L, Liu L, et al. Dynamic alterations of genome and transcriptome in KRAS G13D mutant CRC PDX model treated with cetuximab. BMC Cancer. 2020;20:416. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122. Guo S, Chen D, Huang X, Cai J, Wery JP, Li QX. Cetuximab response in CRC patient‐derived xenografts seems predicted by an expression based RAS pathway signature. Oncotarget. 2016;7:50575‐50581. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123. O'Farrell AC, Jarzabek MA, Lindner AU, et al. Implementing systems modelling and molecular imaging to predict the efficacy of BCL‐2 inhibition in colorectal cancer patient‐derived xenograft models. Cancers (Basel). 2020;12:2978. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124. Zhang Y, Wang X, Qin X, et al. PP2AC level determines differential programming of p38‐TSC‐mTOR signaling and therapeutic response to p38‐targeted therapy in colorectal cancer. EBioMedicine. 2015;2:1944‐1956. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125. Thota R, Yang M, Pflieger L, et al. APC and TP53 mutations predict Cetuximab sensitivity across consensus molecular subtypes. Cancers (Basel). 2021;13:5394. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126. Chen RX, Chen X, Xia LP, et al. N(6)‐methyladenosine modification of circNSUN2 facilitates cytoplasmic export and stabilizes HMGA2 to promote colorectal liver metastasis. Nat Commun. 2019;10:4695. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127. Li T, Hu PS, Zuo Z, et al. METTL3 facilitates tumor progression via an m(6)A‐IGF2BP2‐dependent mechanism in colorectal carcinoma. Mol Cancer. 2019;18:112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128. Wang Y, Lu JH, Wu QN, et al. LncRNA LINRIS stabilizes IGF2BP2 and promotes the aerobic glycolysis in colorectal cancer. Mol Cancer. 2019;18:174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129. Yamaguchi N, Weinberg EM, Nguyen A, et al. PCK1 and DHODH drive colorectal cancer liver metastatic colonization and hypoxic growth by promoting nucleotide synthesis. Elife. 2019;8:e52135. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130. Ni W, Mo H, Liu Y, et al. Targeting cholesterol biosynthesis promotes anti‐tumor immunity by inhibiting long noncoding RNA SNHG29‐mediated YAP activation. Mol Ther. 2021;29:2995‐3010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131. Ma YN, Hong YG, Yu GY, et al. LncRNA LBX2‐AS1 promotes colorectal cancer progression and 5‐fluorouracil resistance. Cancer Cell Int. 2021;21:501. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 132. Liu T, Liu B, Liu Y, et al. Vesicle transporter GOLT1B mediates the cell membrane localization of DVL2 and PD‐L2 and promotes colorectal cancer metastasis. Cancer Cell Int. 2021;21:287. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133. Chen ZH, Qi JJ, Wu QN, et al. Eukaryotic initiation factor 4A2 promotes experimental metastasis and oxaliplatin resistance in colorectal cancer. J Exp Clin Cancer Res. 2019;38:196. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134. Sun H, Meng Q, Shi C, et al. Hypoxia‐inducible exosomes facilitate liver‐tropic Premetastatic niche in colorectal cancer. Hepatology. 2021;74:2633‐2651. [DOI] [PubMed] [Google Scholar]
- 135. Meng X, Peng J, Xie X, et al. Roles of lncRNA LVBU in regulating urea cycle/polyamine synthesis axis to promote colorectal carcinoma progression. Oncogene. 2022;41:4231‐4243. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136. Yao YM, Donoho GP, Iversen PW, et al. Mouse PDX trial suggests synergy of concurrent inhibition of RAF and EGFR in colorectal cancer with BRAF or KRAS mutations. Clin Cancer Res. 2017;23:5547‐5560. [DOI] [PubMed] [Google Scholar]
- 137. Linnekamp JF, Hooff SRV, Prasetyanti PR, et al. Consensus molecular subtypes of colorectal cancer are recapitulated in in vitro and in vivo models. Cell Death Differ. 2018;25:616‐633. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138. Abdirahman SM, Christie M, Preaudet A, et al. A biobank of colorectal cancer patient‐derived xenografts. Cancers (Basel). 2020;12:2340. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139. Burgenske DM, Monsma DJ, Dylewski D, et al. Establishment of genetically diverse patient‐derived xenografts of colorectal cancer. Am J Cancer Res. 2014;4:824‐837. [PMC free article] [PubMed] [Google Scholar]
- 140. Lu M, Sanderson SM, Zessin A, et al. Exercise inhibits tumor growth and central carbon metabolism in patient‐derived xenograft models of colorectal cancer. Cancer Metab. 2018;6:14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141. Guinney J, Dienstmann R, Wang X, et al. The consensus molecular subtypes of colorectal cancer. Nat Med. 2015;21:1350‐1356. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142. Calon A, Lonardo E, Berenguer‐Llergo A, et al. Stromal gene expression defines poor‐prognosis subtypes in colorectal cancer. Nat Genet. 2015;47:320‐329. [DOI] [PubMed] [Google Scholar]
- 143. Isella C, Terrasi A, Bellomo SE, et al. Stromal contribution to the colorectal cancer transcriptome. Nat Genet. 2015;47:312‐319. [DOI] [PubMed] [Google Scholar]
- 144. Straussman R, Morikawa T, Shee K, et al. Tumour micro‐environment elicits innate resistance to RAF inhibitors through HGF secretion. Nature. 2012;487:500‐504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145. Song N, Pogue‐Geile KL, Gavin PG, et al. Clinical outcome from Oxaliplatin treatment in stage II/III colon cancer according to intrinsic subtypes: secondary analysis of NSABP C‐07/NRG oncology randomized clinical trial. JAMA Oncol. 2016;2:1162‐1169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146. Clohessy JG, Pandolfi PP. Mouse hospital and co‐clinical trial project‐‐from bench to bedside. Nat Rev Clin Oncol. 2015;12:491‐498. [DOI] [PubMed] [Google Scholar]
- 147. Izumchenko E, Paz K, Ciznadija D, et al. Patient‐derived xenografts effectively capture responses to oncology therapy in a heterogeneous cohort of patients with solid tumors. Ann Oncol. 2017;28:2595‐2605. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148. Ben‐David U, Ha G, Tseng YY, et al. Patient‐derived xenografts undergo mouse‐specific tumor evolution. Nat Genet. 2017;49:1567‐1575. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149. Pauli C, Hopkins BD, Prandi D, et al. Personalized In vitro and In vivo cancer models to guide precision medicine. Cancer Discov. 2017;7:462‐477. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150. Rusert JM, Juarez EF, Brabetz S, et al. Functional precision medicine identifies new therapeutic candidates for Medulloblastoma. Cancer Res. 2020;80:5393‐5407. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151. Katsiampoura A, Raghav K, Jiang ZQ, et al. Modeling of patient‐derived xenografts in colorectal cancer. Mol Cancer Ther. 2017;16:1435‐1442. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152. Gock M, Kuhn F, Mullins CS, et al. Tumor take rate optimization for colorectal carcinoma patient‐derived xenograft models. Biomed Res Int. 2016;2016:1715053. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153. Julien S, Merino‐Trigo A, Lacroix L, et al. Characterization of a large panel of patient‐derived tumor xenografts representing the clinical heterogeneity of human colorectal cancer. Clin Cancer Res. 2012;18:5314‐5328. [DOI] [PubMed] [Google Scholar]
- 154. Heo EJ, Cho YJ, Cho WC, et al. Patient‐derived xenograft models of epithelial ovarian cancer for preclinical studies. Cancer Res Treat. 2017;49:915‐926. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155. DeRose YS, Wang G, Lin YC, et al. Tumor grafts derived from women with breast cancer authentically reflect tumor pathology, growth, metastasis and disease outcomes. Nat Med. 2011;17:1514‐1520. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156. Siraj S, Masoodi T, Siraj AK, et al. Clonal evolution and timing of metastatic colorectal cancer. Cancers (Basel). 2020;12:2938. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157. Punt CJ, Koopman M, Vermeulen L. From tumour heterogeneity to advances in precision treatment of colorectal cancer. Nat Rev Clin Oncol. 2017;14:235‐246. [DOI] [PubMed] [Google Scholar]
- 158. Zhang Y, Song J, Zhao Z, et al. Single‐cell transcriptome analysis reveals tumor immune microenvironment heterogenicity and granulocytes enrichment in colorectal cancer liver metastases. Cancer Lett. 2020;470:84‐94. [DOI] [PubMed] [Google Scholar]
- 159. Delitto D, Pham K, Vlada AC, et al. Patient‐derived xenograft models for pancreatic adenocarcinoma demonstrate retention of tumor morphology through incorporation of murine stromal elements. Am J Pathol. 2015;185:1297‐1303. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160. Chao C, Widen SG, Wood TG, et al. Patient‐derived xenografts from colorectal carcinoma: a temporal and hierarchical study of murine stromal cell replacement. Anticancer Res. 2017;37:3405‐3412. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161. Liu WN, Fong SY, Tan WWS, et al. Establishment and characterization of humanized mouse NPC‐PDX model for testing immunotherapy. Cancers (Basel). 2020;12:1025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162. Wang M, Yao LC, Cheng M, et al. Humanized mice in studying efficacy and mechanisms of PD‐1‐targeted cancer immunotherapy. FASEB J. 2018;32:1537‐1549. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163. Seebacher NA, Stacy AE, Porter GM, Merlot AM. Clinical development of targeted and immune based anti‐cancer therapies. J Exp Clin Cancer Res. 2019;38:156. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164. Doleschel D, Hoff S, Koletnik S, et al. Regorafenib enhances anti‐PD1 immunotherapy efficacy in murine colorectal cancers and their combination prevents tumor regrowth. J Exp Clin Cancer Res. 2021;40:288. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165. Singh SP, Singh R, Gupta OP, Gupta S, Brahma Bhatt ML. Immunotherapy: newer therapeutic armamentarium against cancer stem cells. J Oncol. 2020;2020:3963561. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166. Chicote I, Camara JA, Palmer HG. Advanced colorectal cancer Orthotopic patient‐derived xenograft models for cancer and stem cell research. Methods Mol Biol. 2020;2171:321‐329. [DOI] [PubMed] [Google Scholar]
- 167. Morton CL, Houghton PJ. Establishment of human tumor xenografts in immunodeficient mice. Nat Protoc. 2007;2:247‐250. [DOI] [PubMed] [Google Scholar]
- 168. Drost J, Clevers H. Organoids in cancer research. Nat Rev Cancer. 2018;18:407‐418. [DOI] [PubMed] [Google Scholar]
- 169. Jacob F, Salinas RD, Zhang DY, et al. A patient‐derived glioblastoma organoid model and biobank recapitulates inter‐ and intra‐tumoral heterogeneity. Cell. 2020;180:188‐204.e22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170. Pasch CA, Favreau PF, Yueh AE, et al. Patient‐derived cancer organoid cultures to predict sensitivity to chemotherapy and radiation. Clin Cancer Res. 2019;25:5376‐5387. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171. Driehuis E, Kretzschmar K, Clevers H. Establishment of patient‐derived cancer organoids for drug‐screening applications. Nat Protoc. 2020;15:3380‐3409. [DOI] [PubMed] [Google Scholar]
- 172. Lee SH, Hu W, Matulay JT, et al. Tumor evolution and drug response in patient‐derived organoid models of bladder cancer. Cell. 2018;173:515‐528.e17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173. Xu Y, Chen J, Huang Y, et al. Patient‐derived organoids in cellulosic sponge model chemotherapy response of metastatic colorectal cancer. Clin Transl Med. 2021;11:e285. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174. Li H, Dai W, Xia X, et al. Modeling tumor development and metastasis using paired organoids derived from patients with colorectal cancer liver metastases. J Hematol Oncol. 2020;13:119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175. Codrich M, Dalla E, Mio C, et al. Integrated multi‐omics analyses on patient‐derived CRC organoids highlight altered molecular pathways in colorectal cancer progression involving PTEN. J Exp Clin Cancer Res. 2021;40:198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176. Yao Y, Xu X, Yang L, et al. Patient‐derived organoids predict Chemoradiation responses of locally advanced rectal cancer. Cell Stem Cell. 2020;26:17‐26.e6. [DOI] [PubMed] [Google Scholar]
- 177. Weeber F, van de Wetering M, Hoogstraat M, et al. Preserved genetic diversity in organoids cultured from biopsies of human colorectal cancer metastases. Proc Natl Acad Sci U S A. 2015;112:13308‐13311. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178. Schutte M, Risch T, Abdavi‐Azar N, et al. Molecular dissection of colorectal cancer in pre‐clinical models identifies biomarkers predicting sensitivity to EGFR inhibitors. Nat Commun. 2017;8:14262. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179. Ganesh K, Wu C, O'Rourke KP, et al. A rectal cancer organoid platform to study individual responses to chemoradiation. Nat Med. 2019;25:1607‐1614. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180. Ooft SN, Weeber F, Dijkstra KK, et al. Patient‐derived organoids can predict response to chemotherapy in metastatic colorectal cancer patients. Sci Transl Med. 2019;11:eaay2574. [DOI] [PubMed] [Google Scholar]
- 181. Townsend EC, Murakami MA, Christodoulou A, et al. The public repository of xenografts enables discovery and randomized phase II‐like trials in mice. Cancer Cell. 2016;29:574‐586. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182. Bruna A, Rueda OM, Greenwood W, et al. A biobank of breast cancer explants with preserved intra‐tumor heterogeneity to screen anticancer compounds. Cell. 2016;167:260‐274.e22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183. Teng S, Li YE, Yang M, et al. Tissue‐specific transcription reprogramming promotes liver metastasis of colorectal cancer. Cell Res. 2020;30:34‐49. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184. Guo J, Xu G, Mao C, Wei R. Low expression of Smurf1 enhances the Chemosensitivity of human colorectal cancer to gemcitabine and cisplatin in patient‐derived xenograft models. Transl Oncol. 2020;13:100804. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185. Ji X, Chen S, Guo Y, et al. Establishment and evaluation of four different types of patient‐derived xenograft models. Cancer Cell Int. 2017;17:122. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186. Chen D, Huang X, Cai J, et al. A set of defined oncogenic mutation alleles seems to better predict the response to cetuximab in CRC patient‐derived xenograft than KRAS 12/13 mutations. Oncotarget. 2015;6:40815‐40821. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187. Chen I, Lorentzen T, Linnemann D, et al. Seeding after ultrasound‐guided percutaneous biopsy of liver metastases in patients with colorectal or breast cancer. Acta Oncol. 2016;55:638‐643. [DOI] [PubMed] [Google Scholar]
- 188. Kageyama K, Ohara M, Saito K, et al. Establishment of an orthotopic patient‐derived xenograft mouse model using uveal melanoma hepatic metastasis. J Transl Med. 2017;15:145. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189. Leong TL, Marini KD, Rossello FJ, et al. Genomic characterisation of small cell lung cancer patient‐derived xenografts generated from endobronchial ultrasound‐guided transbronchial needle aspiration specimens. PLoS One. 2014;9:e106862. [DOI] [PMC free article] [PubMed] [Google Scholar]
