Next Article in Journal
Orphan Nuclear Receptor Nur77 Mediates the Lethal Endoplasmic Reticulum Stress and Therapeutic Efficacy of Cryptomeridiol in Hepatocellular Carcinoma
Next Article in Special Issue
CRC Therapy Identifies Indian Hedgehog Signaling in Mouse Endometrial Epithelial Cells and Inhibition of Ihh-KLF9 as a Novel Strategy for Treating IUA
Previous Article in Journal
Differentially Expressed Cell Cycle Genes and STAT1/3-Driven Multiple Cancer Entanglement in Psoriasis, Coupled with Other Comorbidities
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Translation Potential and Challenges of In Vitro and Murine Models in Cancer Clinic

1
Shanghai LIDE Biotech Co., Ltd., Shanghai 201203, China
2
China Innovation Center of Roche, Roche R & D Center, Shanghai 201203, China
*
Authors to whom correspondence should be addressed.
Cells 2022, 11(23), 3868; https://doi.org/10.3390/cells11233868
Submission received: 9 October 2022 / Revised: 20 November 2022 / Accepted: 23 November 2022 / Published: 30 November 2022
(This article belongs to the Special Issue Reprogrammed Cells in Disease Modeling and Drug Discovery II)

Abstract

:
As one of the leading causes of death from disease, cancer continues to pose a serious threat to human health globally. Despite the development of novel therapeutic regimens and drugs, the long-term survival of cancer patients is still very low, especially for those whose diagnosis is not caught early enough. Meanwhile, our understanding of tumorigenesis is still limited. Suitable research models are essential tools for exploring cancer mechanisms and treatments. Herein we review and compare several widely used in vitro and in vivo murine cancer models, including syngeneic tumor models, genetically engineered mouse models (GEMM), cell line-derived xenografts (CDX), patient-derived xenografts (PDX), conditionally reprogrammed (CR) cells, organoids, and MiniPDX. We will summarize the methodology and feasibility of various models in terms of their advantages and limitations in the application prospects for drug discovery and development and precision medicine.

1. Introduction

In the past few decades, driven by breakthroughs in cancer research, the number of anti-cancer drugs has soared [1]. Consequently, cancer patients are living longer, yet mortality and morbidity in the vast majority of cancer patients are still devastating [2]. More recently, despite remarkable progress in the research of targeted anti-cancer drugs and immunotherapies, there are still challenges associated with drug efficacy, toxicity, and resistance. Thus, the urgency remains for more transformative treatments in order to address these unmet medical needs. Unfortunately, in contrast to drugs for the treatment of other diseases, the likelihood for a new phase one oncology clinical compound to reach market approval is significantly lower [3,4]. One reason for the low success rate is the translatability of preclinical cancer models to patients in clinic. Not only do the defects of the research model limit the development of new drugs, they also hinder the progress of precision medicine [5,6]. So far, genetic, epigenetic, and environmental factors have been frequently invoked to account for the development of tumors. At the same time, the pathogenesis of cancer is yet to be further elucidated. Our understanding of disease biology is still not always sufficient to effectively prevent or accurately predict tumor progression. As such, cancer models that can mimic tumor genesis and development are helpful for mechanistic research on cancer and could potentially enable better treatment options for cancer patients. In this review, we aim to discuss the features, utility, advantages, and disadvantages of various preclinical cancer models.

2. Syngeneic Tumor Models

Syngeneic tumor models are established by subcutaneously or orthotopically injecting mouse tumor cell lines expanded in vitro into immunocompetent mice. Syngeneic tumor models have been used in cancer research and drug development for more than 50 years across various cancer types, including lung, breast, bowel, liver, and malignant melanoma [7,8,9]. Established in immunocompetent mice, the syngeneic models are therefore suitable for evaluating drugs that affect immunity. Mosely et al. examined the responses of anti-CTLA-4 and anti-PD-L1 therapies in a panel of commonly used murine syngeneic tumor models, including 4T1, MC38, B16F10 AP-3, CT26, and LL/2. These syngeneic tumor models displayed variable responsiveness to immunotherapies, with significant differential gene expression panels identified, correlating to the tumor immune infiltrates. These results suggest there are two phenotypes of models, including “inflamed” and “non-inflamed”. Choosing a suitable model is crucial for the research of immunotherapies [10]. Since cell lines can be expanded rapidly and reproducibly in vitro, syngeneic mouse models have the advantages of being relatively fast to establish, low-cost, and reproducible. However, the use of cell lines also leads to a lack of genomic, epigenetic, and micro-environmental heterogeneity in this model, which cannot mimic the complex situations of patients well [11]. Another disadvantage is that only a few cell lines can be harnessed to establish syngeneic mouse models, thereby limiting the applications of these models.

3. Genetically Engineered Mouse Models (GEMM)

Genetically engineered mouse models are autochthonous cancer models driven by the expression of oncogenes or deletion of tumor suppressors using genetic engineering technology. Research on oncogenes and the development of genetic engineering technology contributed to the invention of genetically engineered mice. These models primarily use tissue-specific promoters to drive oncogene expression or tissue-specific recombinases to drive the loss of tumor suppressors [12,13,14,15,16]. The genetically engineered mouse model is a spontaneous model that can mimic the entire precancerous lesion and tumorigenesis processes. Therefore, this model has been applied for biomarker development [17,18], cancer mechanism research, and preclinical drug testing [19,20,21]. For example, the transgenic adenocarcinoma of the mouse prostate (TRAMP) is a model of prostate cancer hormonally regulated by androgens. This model develops progressive, multifocal, and heterogeneous prostate cancer, correlating with sexual maturity, which closely mimics the clinical disease [19]. At the same time, a genetically engineered mouse model is also suitable for studying immunotherapy and immune-related drugs. Genetically engineered mouse models have several advantages, such as being autochthonous and carrying an intact immune system and microenvironment. However, cancer models caused by alterations in a tumor-driver gene do not fully mimic the complexity of human cancer mechanisms. Meanwhile, the tumor formation in GEMM is variable and has longer latency periods than transplantable models, thus requiring large colonies of mice and extended experimental timelines. In addition, only a handful of genetically engineered mouse models are available, thus limiting the use of this technology in cancer research.

4. Cell Lines and Cell Line-Derived Xenograft (CDX)

Traditional cancer cell lines as tools used in vitro and in vivo have made significant contributions to cancer research and drug discovery and development for decades [22]. A large amount of the original cell lines were established by the National Cancer Institute (NCI) and the Hamon Center for Therapeutic Oncology Research. In addition to the American Type Culture Collection (ATCC), cell lines from cell banks all over the world can now be utilized by researchers [23]. Like syngeneic tumor models, cell line-derived xenograft (CDX) is also established by injecting mouse tumor cell lines expanded in vitro into mice. With the advantages of good reproducibility, rapid turn-around time, common availability, and cost-efficient generation, it is clear why CDX models are widely used in efficacy, pharmacokinetics (PK), and pharmacodynamics (PD) studies in drug discovery [24,25]. Among numerous examples, Suzawa et al. examined the antitumor activity of afatinib, an irreversible epidermal growth factor receptor (EGFR)-HER2 dual inhibitor, in several non-small cell lung cancer (NSCLC) cell lines, including A549, Calu-3, HCC827, NCI-H1299, NCI-H1781, NCI-H1975, NCIH1993, and NCI-H2170, to explore the correlation between the genetic alterations of cells and cell sensitivity to afatinib. The HER2-altered cell lines (H2170, Calu-3, and H1781) were found to be sensitive to afatinib, while the HER2- or EGFR-non-dependent NSCLC cells were insensitive, which implied that afatinib was a potential option for NSCLC patients with HER2 alterations [24].
However, CDX models have several significant shortcomings. First, the biological characteristics of cells in vitro for long-term passage may change. This leads to differences in tumorigenicity and the genomic and epigenetic properties of cell lines used in different laboratories. Second, a cell-line-derived xenograft has low heterogeneity compared to the original tumor. Third, CDXs are established in immune-deficient mice, thus limiting the application in the study of immunotherapy and immunomodulatory drugs [26,27,28,29,30].

5. Patient-Derived Xenografts (PDX)

Patient-derived xenografts (PDX) are established by subcutaneous or orthotopic implantation of surgical or biopsy tissue chunks into immune-deficient mice (Figure 1C) [31,32]. Tumor cells enriched from hydrothorax, ascites, and circulating tumor cells can also be used for PDX establishment [33,34,35]. After implantation, PDX requires a long latency period before tumor formation. The latency period can range from a couple of weeks to several months, depending on the type of cancer, the modeling method, and the growth characteristics of the original tumor [36]. PDX can be used for long-term preservation and pharmacodynamic studies after several cycles of serial in vivo transplantation [37]. At present, PDX has been successfully established in various cancers, including lung cancer [38], breast cancer [39,40], pancreatic cancer [41], colon and rectal cancer [42,43], gastric cancer [44], ovarian cancer [45], hepatocellular carcinoma [46], prostate cancer [47], brain tumors [48], melanoma [49], and head and neck tumors [50], etc. The success rates of modeling vary widely among cancer types from 10–100% as reported. A number of studies have proven that PDX can maintain the genetic profile, gene expression patterns, tissue histology, and drug response of the original tumor, as well as the molecular and cellular heterogeneity of the primary tumor [42,51]. In addition, PDX models also exhibit the possibility of predicting metastatic potential and drug response [52]. Currently, PDX models are widely considered a more physiologically relevant preclinical model than CDX and are widely used in the research of cancer mechanisms and the discovery of anti-tumor drugs [53,54,55]. Gao et al. established ~1000 patient-derived tumor xenograft models with multiple driver gene mutations to evaluate the population responses to 62 treatments from six cancer indications. Correlation between genotypes and drug response was identified from the research, showcasing the impressive progress in elucidating mechanisms of drug resistance and the profiling of therapeutic candidates [53].
Despite its significant advantages, PDX is less suited for large-scale drug screening and personalized precision medicine due to its long turn-around time and high cost. Usually, a 6–8-month period is required for tumor xenograft engraftment and efficacy studies [56,57,58]. Established in immune deficient mice, PDX models are not suitable for evaluating the efficacy of immunomodulators. In addition, one study showed that human stromal cells in PDX decreased gradually over passages, and were gradually replaced by mouse cells [59].

6. Conditionally Reprogrammed (CR) Cells

In addition to in vivo models, in vitro models, such as traditional cell lines, are also important tools for cancer research. However, primary tumor cells are difficult to proliferate in vitro, and the success rate of establishing cancer cell lines is as low as 1–10% [60,61]. Conditional reprogramming technology (CRT) effectively improves the success rate of primary cell culture and cell line establishment. One critical success factor of CRT is the use of feeder cells and ROCK inhibitors to help tumor cells and other epithelial cells proliferate continuously in vitro (Figure 1A) [62,63,64]. At present, multiple cancer cells have been established using CRT [64], including adenoid cystic carcinoma [65], breast [66], prostate [67,68], pancreatic [69], colorectal [70], lung [71,72], cervical [73], skin, kidney, ovarian cancers, and so on [64,74]. In addition, conditionally reprogrammed cells can be generated from PDX models and organoids [75,76,77] and passaged more than 200 times while preserving the heterogeneity of tumor cells [78,79]. As a suitable in vitro tool, CRT has been applied to drug development and precision medicine [65,68,80,81]. For example, Rei et al. established conditionally reprogrammed cells from patients with recurrent hormone receptor-positive (HR+)/human epidermal receptor 2-negative (HER2−) breast cancer to test heterogeneity and drug sensitivity. The mutation status and pathological features were preserved in CR cells, while the RNA expression was different from the primary tumor cells. The Rei group tested the responses of CR cells derived from an ER+/PgR+/HER2− liver metastasis to 224 drugs, and 66 compounds reduced cell viability, including SERD and CDK4/6 inhibitors. The original patient used SERD and CDK4/6 inhibitors after metastasectomy, and no recurrence was noted for 13 months, consistent with drug screening results on CR cells [80]. In another research project in prostate cancer, researchers generated CR cells from seven patients with diseases ranging from primary prostate cancer to advanced castration-resistant prostate cancer (CRPC). A high-throughput drug screening of 306 emerging and clinical cancer drugs led to the identification of navitoclax, an inhibitor of the Bcl-2 family, as the most effective drug against CRPC CR cells. In addition, taxanes, mepacrine, and retinoids were also proven potent in the screening [68].
Not only can CR cells be directly maintained in 2D culture, but they may also be grown into spheroids or organoids [67]. CR cells can also be inoculated into mice to obtain PDX models [62,63], which provides a new approach to establishing better performing models, including those for breast cancer and other cancers that have had low success rates in PDX.
One major limitation of CR cells or any in vitro model is that they are incapable of capturing the PK/PD properties of the compounds being evaluated. The concentration of the compounds being tested is typically constant, which is very different from the dynamic drug concentration in vivo due to the drug’s absorption, distribution, metabolism, and excretion (ADME) processes. Furthermore, in vitro cancer models are, in general, not suitable for the evaluation of prodrugs. For example, as a prodrug of 5-FU (5-fluorouracil), tegafur is slowly metabolized to 5-FU in the liver, thereby leading to lower toxicities than 5-FU [82]. Tegafur is clinically widely used in the treatment of numerous tumors [83,84], but its anti-tumor effect cannot be assessed in vitro directly in the absence of metabolizing enzymes.

7. Patient-Derived Organoid

Organoids are the organotypic structures established in vitro in 3D. The first adult stem cell-derived organoids were established from Lgr5-expressing mouse intestinal stem cells [85]. In recent years, organoid technology has been applied to the generation of organoids derived from cells isolated from patient cancer tissues and circulating tumor cells (Figure 1B) [86,87]. Organoids derived from patients can proliferate in vitro and be maintained in long-term cryopreservation. Besides the proliferation ability, organoids could also capture the heterogeneity of cancer.
Oded et al. established 56 organoid cultures from 32 patients, representing all main subtypes of ovarian cancer. The organoids recapitulated the hallmarks and tumor heterogeneity of ovarian cancer and demonstrated intra- and inter-patient heterogeneity. These organoids were also used for drug-screening assays to evaluate tumor subtype responses to chemotherapy [88]. Compared with PDX, organoids require a shorter experimental period and a lower cost, which can be used for medium–high throughput drug screening. At the same time, the establishment of patient-derived organoids allows the evaluation of anti-cancer drugs as an approach to identifying precision medicines. Pauli et al. collected 145 specimens of 18 different tumor types derived from 769 patients and subsequently established 56 patient-derived organoid cultures, which were used in high-throughput drug screening to discover effective treatment options [89]. It should be noted that several translational studies on organoids and precision medicine have delivered promising results [90,91,92,93,94]. For example, in the research of pancreatic cancer, patient-derived cancer organoids exhibited responses to chemotherapeutics, consistent with the efficacy observed in patients, and contributed to the evaluation of synchronous metastases. Based on the gene expression signatures of organoids, responses to chemotherapy and targeted drugs could be predicted [93]. In addition, Tiriac et al. collected 112 biopsy tissues of locally advanced rectal cancer (LARC) from patients enrolled in a phase III clinical trial of neoadjuvant chemoradiation (NACR) and established 96 patient-derived organoids (PDOs). Responses to radiation and chemotherapy on PDOs were consistent with patients’ clinical responses, with 84.43% accuracy, 78.01% sensitivity, and 91.97% specificity [94]. Organoids can also be engrafted, enabling in vivo model establishment and in vivo drug efficacy studies [88].
Although organoids are rapid and cost-effective, several shortcomings may limit their application in cancer research. The cell population of an organoid will gradually decrease during the culture process, and the heterogeneity is lower compared to PDX. In addition, it can only be administered in vitro and, hence, is not suitable for evaluating molecules in the context of systemic administration, metabolism, and distribution [61].

8. MiniPDX

In 2015, researchers proposed the concept of “next-generation functional diagnostics” [95] similar to next-generation sequencing (NGS), including 2D ex vivo drug toxicity testing, patient-derived xenografts (PDX), 3D organoids, and other technologies. The core purpose of these functional tests was to address the limitations of genome-based cancer therapeutic matching. MiniPDX, as an in vivo version of organoid technology, was developed to provide a more rapid and accurate drug efficacy test to guide personalized cancer treatments [96]. Fresh tumor specimens acquired from patients, including tumor tissues obtained from surgery, biopsy, puncture, pleural fluid, and ascites, are digested and purified to prepare cell suspensions. Then, the cell suspensions are filled into the MiniPDX capsules. After subcutaneous implantation, immunodeficient mouse-bearing capsules are treated for 7 days with candidate drugs or their combinations. Post-treatment, the implanted MiniPDX capsules are retrieved, and the relative luminance unit (RLU) is examined for tumor cell proliferation assessment. The calculated tumor cell growth inhibition (TCGI) (%) reflects the PDX response to treatment options in this model (Figure 1D).
The MiniPDX cancer drug sensitivity test has several favorable features, such as a short cycle time and high consistency, which are typically aligned with results from in vivo drug sensitivity tests and clinical treatment. MiniPDX drug sensitivity tests generate results in about 10 days, which can not only meet the requirements of preclinical research of antitumor drugs but also enable fast and accurate drug sensitivity results for patients in a clinical setting. In China, the MiniPDX drug sensitivity test model has benefited numerous tumor patients. For example, a patient with endometrial stromal sarcoma had lung metastasis two chemotherapy cycles after operation. The metastatic lesions were taken for a MiniPDX drug sensitivity test. It was found that the patient was sensitive to apatinib alone and apatinib combined with olaparib. Four months after changing the medication regimen, the patient’s lung metastasis began to subside [96]. In another example, the chemotherapy regimen supported by a MiniPDX drug sensitivity test significantly prolonged the survival time of patients with gallbladder cancer, with the median overall survival time increasing from 13.9 months to 18.6 months and the disease-free progression time increased from 12 months to 17.6 months [97]. Another study of metastatic duodenal adenocarcinoma combined MiniPDX with NGS to reveal the mutations of key genes while conducting rapid drug sensitivity tests to provide individualized treatment for patients [98]. In a study on patients with platinum-resistant ovarian cancer, the clinical treatment response rate guided by MiniPDX drug sensitivity detection can be as high as 75% [99]. To treat patients with non-small cell lung cancer, Chen et al. confirmed that the MiniPDX-guided treatment group showed a better OS and PFS than the conventional chemotherapy group [100]. Li and colleagues verified the drug response predicted by WES, proteomics, and phosphorproteomics on the MiniPDX model and established an entire workflow from the generation of large omics datasets to in vivo drug testing models of colorectal cancer [101]. MiniPDX drug sensitivity test results were largely consistent with clinical responses in more and more patients with different tumor types [102,103], which indicated that MiniPDX models have great potential for guiding personalized cancer therapy.
Although the MiniPDX drug sensitivity test model has made significant progress in clinical practice, some limitations exist, impacting the application and wide adoption of the MiniPDX model. For example, the MiniPDX drug sensitivity test model uses immunodeficient mice and thus cannot be used for the detection of antitumor immunotherapeutic drugs. In addition, animal rooms and cell culture rooms meeting specific regulations and qualifications are required to complete the MiniPDX test. Finally, due to the use of immunodeficient mice and the high operation and maintenance costs of animal experimental facilities, the cost of a MiniPDX drug sensitivity test is higher than that of a typical in vitro drug sensitivity test.

9. Conclusions and Perspective

There are various models for cancer research and anti-tumor drug discovery and development. The experimental methods and underlying mechanisms of these models are different (Figure 1), and each has its own advantages and disadvantages (Table 1). It is critical to select the most appropriate model in order to meet the needs of different research purposes. In vitro models, such as CRT and organoids, are relatively fast, cost-effective, and suitable for high-throughput screening. However, they are only suitable for evaluating certain types of drugs because of the absence of therapeutically relevant administration and PK/PD correlation.
Among the various in vivo models, syngeneic tumor models and GEMM are suitable for immune oncology studies, but gaps between the physiological characteristics of humans and mice limit their application. The CDX model is derived from human tumor cell lines, but it lacks heterogeneity due to the limited types of traditional cell lines. PDX has previously shown a high degree of translatability to patients, and heterogeneity after continuous passage can be maintained. However, due to its long turnaround time, PDX is more suitable for drug development than precision medicine. With the advantages of rapidness, cost-effectiveness, a wide application range, and a high success rate, MiniPDX is expected to become an important tool in precision medicine and anti-tumor drug research.
As anticancer drug discovery and development continue to evolve, the applications of the aforementioned models and their future refinement will play an important role in giving drug hunters and developers the evidence and confidence to advance molecules to clinical studies. This also applies for physicians making informed medication decisions. Even for the same research program and oncology target, data from multiple in vitro and in vivo models could corroborate, and help assess the potential of therapeutic intervention from different angles. The selection of cancer in vitro and in vivo models and the interpretation of data rely on a solid understanding of the study methods and the understanding of the advantages and limitations of these models.

10. Disclosure

Yuan Long and Bin Xie are employed by Shanghai LIDE Biotech Co., Ltd. Danyi Wen is the CEO and founder of Shanghai LIDE Biotech Co., Ltd., and she is an inventor of the patent application named “METHOD AND DEVICE FOR DRUG SCREENING”, filed by Shanghai LIDE Biotech Co., Ltd. The patent application inventors are Danyi Wen and Feifei Zhang. Hong C. Shen declares no conflicts of interest.

Funding

This study was supported by grants from Natural Science Foundation of Liaoning Province, China (2019-MS-375).

Acknowledgments

We appreciate Josh Caggiula and Loc Van for their help in language correction.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Sengupta, R.; Zaidi, S.K. Discovery Science Driving Clinical Breakthroughs. Clin. Cancer Res. 2021, 27, 5757–5759. [Google Scholar] [CrossRef]
  2. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. GLO-BOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef]
  3. Rubin, E.H.; Gilliland, D.G. Drug development and clinical trials—The path to an approved cancer drug. Nat. Rev. Clin. Oncol. 2012, 9, 215–222. [Google Scholar] [CrossRef]
  4. Hay, M.; Thomas, D.W.; Craighead, J.L.; Economides, C.; Rosenthal, J. Clinical development success rates for investigational drugs. Nat. Biotechnol. 2014, 32, 40–51. [Google Scholar] [CrossRef] [PubMed]
  5. Boehm, J.; Golub, T.R. An ecosystem of cancer cell line factories to support a cancer dependency map. Nat. Rev. Genet. 2015, 16, 373–374. [Google Scholar] [CrossRef] [PubMed]
  6. Codenotti, S.; Mansoury, W.; Pinardi, L.; Monti, E.; Marampon, F.; Fanzani, A. Animal models of well-differentiated/dedifferentiated liposarcoma: Utility and limitations. OncoTargets Ther. 2019, 12, 5257–5268. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Abolhassani, M.; Guais, A.; Sanders, E.; Campion, F.; Fichtner, I.; Bonte, J.; Baronzio, G.; Fiorentini, G.; Israël, M.; Schwartz, L. Screening of well-established drugs targeting cancer metabolism: Reproducibility of the efficacy of a highly effective drug combination in mice. Investig. New Drugs 2011, 30, 1331–1342. [Google Scholar] [CrossRef]
  8. Vallespi, M.G.; Pimentel, G.; Cabrales-Rico, A.; Garza, J.; Oliva, B.; Mendoza, O.; Gomez, Y.; Basaco, T.; Sánchez, I.; Calderon, C.; et al. Antitumor efficacy, pharmacokinetic and biodistribution studies of the anticancer peptide CIGB-552 in mouse models. J. Pept. Sci. 2014, 20, 850–859. [Google Scholar] [CrossRef]
  9. Behrens, D.; Walther, W.; Fichtner, I. Pancreatic cancer models for translational research. Pharmacol. Ther. 2017, 173, 146–158. [Google Scholar] [CrossRef] [PubMed]
  10. Mosely, S.I.; Prime, J.E.; Sainson, R.C.; Koopmann, J.-O.; Wang, D.Y.; Greenawalt, D.M.; Ahdesmaki, M.J.; Leyland, R.; Mullins, S.; Pacelli, L.; et al. Rational Selection of Syngeneic Preclinical Tumor Models for Immunotherapeutic Drug Discovery. Cancer Immunol. Res. 2017, 5, 29–41. [Google Scholar] [CrossRef] [PubMed]
  11. Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Greenberg, N.M.; DeMayo, F.; Finegold, M.J.; Medina, D.; Tilley, W.D.; Aspinall, J.O.; Cunha, G.R.; Donjacour, A.A.; Matusik, R.J.; Rosen, J.M. Prostate cancer in a transgenic mouse. Proc. Natl. Acad. Sci. USA 1995, 92, 3439–3443. [Google Scholar] [CrossRef] [Green Version]
  13. Sinn, E.; Muller, W.; Pattengale, P.; Tepler, I.; Wallace, R.; Leder, P. Coexpression of MMTV/v-Ha-ras and MMTV/c-myc genes in transgenic mice: Synergistic action of oncogenes in vivo. Cell 1987, 49, 465–475. [Google Scholar] [CrossRef] [PubMed]
  14. Shibata, H.; Toyama, K.; Shioya, H.; Ito, M.; Hirota, M.; Hasegawa, S.; Matsumoto, H.; Takano, H.; Akiyama, T.; Toyoshima, K.; et al. Rapid Colorectal Adenoma Formation Initiated by Conditional Targeting of the Apc Gene. Science 1997, 278, 120–123. [Google Scholar] [CrossRef] [PubMed]
  15. Chen, Z.; Trotman, L.C.; Shaffer, D.; Lin, H.-K.; Dotan, Z.A.; Niki, M.; Koutcher, J.A.; Scher, H.I.; Ludwig, T.; Gerald, W.; et al. Crucial role of p53-dependent cellular senescence in suppression of Pten-deficient tumorigenesis. Nature 2005, 436, 725–730. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Hooijkaas, A.I.; Gadiot, J.; van der Valk, M.; Mooi, W.J.; Blank, C.U. Targeting BRAF in an Inducible Murine Model of Mela-noma. Am. J. Pathol. 2012, 181, 785–794. [Google Scholar] [CrossRef]
  17. Pitteri, S.J.; JeBailey, L.; Faça, V.M.; Thorpe, J.D.; Silva, M.A.; Ireton, R.C.; Horton, M.B.; Wang, H.; Pruitt, L.C.; Zhang, Q.; et al. Integrated Proteomic Analysis of Human Cancer Cells and Plasma from Tumor Bearing Mice for Ovarian Cancer Biomarker Discovery. PLoS ONE 2009, 4, e7916. [Google Scholar] [CrossRef]
  18. Fijneman, R.J.; de Wit, M.; Pourghiasian, M.; Piersma, S.R.; Pham, T.V.; Warmoes, M.O.; Lavaei, M.; Piso, C.; Smit, F.; Diemen, P.M.D.-V.; et al. Proximal Fluid Proteome Profiling of Mouse Colon Tumors Reveals Biomarkers for Early Diagnosis of Human Colorectal Cancer. Clin. Cancer Res. 2012, 18, 2613–2624. [Google Scholar] [CrossRef] [Green Version]
  19. Kaplan-Lefko, P.J.; Chen, T.-M.; Ittmann, M.M.; Barrios, R.J.; Ayala, G.E.; Huss, W.J.; Maddison, L.A.; Foster, B.A.; Greenberg, N.M. Pathobiology of autochthonous prostate cancer in a pre-clinical transgenic mouse model. Prostate 2003, 55, 219–237. [Google Scholar] [CrossRef]
  20. Hingorani, S.R.; Wang, L.; Multani, A.S.; Combs, C.; Deramaudt, T.B.; Hruban, R.H.; Rustgi, A.K.; Chang, S.; Tuveson, D.A. Trp53R172H and KrasG12D cooperate to promote chromosomal instability and widely metastatic pancreatic ductal adeno-carcinoma in mice. Cancer Cell 2005, 7, 469–483. [Google Scholar] [CrossRef]
  21. Kucherlapati, R. Genetically Modified Mouse Models for Biomarker Discovery and Preclinical Drug Testing. Clin. Cancer Res. 2012, 18, 625–630. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Sharma, S.V.; Haber, D.A.; Settleman, J. Cell line-based platforms to evaluate the therapeutic efficacy of candidate anticancer agents. Nat. Cancer 2010, 10, 241–253. [Google Scholar] [CrossRef]
  23. Huo, K.-G.; D’Arcangelo, E.; Tsao, M.-S. Patient-derived cell line, xenograft and organoid models in lung cancer therapy. Transl. Lung Cancer Res. 2020, 9, 2214–2232. [Google Scholar] [CrossRef] [PubMed]
  24. Suzawa, K.; Toyooka, S.; Sakaguchi, M.; Morita, M.; Yamamoto, H.; Tomida, S.; Ohtsuka, T.; Watanabe, M.; Hashida, S.; Maki, Y.; et al. Antitumor effect of afatinib, as a human epidermal growth factor receptor 2-targeted therapy, in lung cancers har-boring HER 2 oncogene alterations. Cancer Sci. 2015, 107, 45–52. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Kasiri, S.; Shao, C.; Chen, B.; Wilson, A.N.; Yenerall, P.; Timmons, B.C.; Girard, L.; Tian, H.; Behrens, C.; Wistuba, I.I.; et al. GLI1 Blockade Potentiates the Antitumor Activity of PI3K Antagonists in Lung Squamous Cell Carcinoma. Cancer Res. 2017, 77, 4448–4459. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Otto, R.; Sers, C.; Leser, U. Robust in-silico identification of cancer cell lines based on next generation sequencing. Oncotarget 2017, 8, 34310–34320. [Google Scholar] [CrossRef] [Green Version]
  27. Vaughan, L.; Glänzel, W.; Korch, C.; Capes-Davis, A. Widespread Use of Misidentified Cell Line KB (HeLa): Incorrect At-tribution and Its Impact Revealed through Mining the Scientific Literature. Cancer Res. 2017, 77, 2784–2788. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Lin, T.J.; Beal, K.M.; DeGruttola, H.S.; Brennan, S.; Marzilli, L.A.; Anderson, K. Utilization of sequence variants as biomarkers to analyze population dynamics in cloned cell lines. Biotechnol. Bioeng. 2017, 114, 1744–1752. [Google Scholar] [CrossRef]
  29. Horbach, S.P.J.M.; Halffman, W. The ghosts of HeLa: How cell line misidentification contaminates the scientific literature. PLoS ONE 2017, 12, e0186281. [Google Scholar] [CrossRef] [Green Version]
  30. Reiter, J.G.; Makohon-Moore, A.P.; Gerold, J.M.; Heyde, A.; Attiyeh, M.A.; Kohutek, Z.A.; Tokheim, C.J.; Brown, A.; DeBlasio, R.M.; Niyazov, J.; et al. Minimal functional driver gene heterogeneity among untreated metastases. Science 2018, 361, 1033–1037. [Google Scholar] [CrossRef]
  31. Perales-Patón, J.; Piñeiro-Yáñez, E.; Tejero, H.; Lopez-Casas, P.P.; Hidalgo, M.; López, G.G.; Al-Shahrour, F. Pancreas Cancer Precision Treatment Using Avatar Mice from a Bioinformatics Perspective. Public Heal. Genom. 2017, 20, 81–91. [Google Scholar] [CrossRef] [PubMed]
  32. Walters, D.M.; Stokes, J.B.; Adair, S.J.; Stelow, E.B.; Borgman, C.A.; Lowrey, B.T.; Xin, W.; Blais, E.M.; Lee, J.K.; Papin, J.A.; et al. Clinical, Molecular and Genetic Validation of a Murine Orthotopic Xenograft Model of Pancreatic Adenocarcinoma Using Fresh Human Specimens. PLoS ONE 2013, 8, e77065. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Lee, A.Q.; Ijiri, M.; Rodriguez, R.; Gandour-Edwards, R.; Lee, J.; Tepper, C.G.; Li, Y.; Beckett, L.; Lam, K.; Goodwin, N.; et al. Novel Patient Metastatic Pleural Effusion-Derived Xenograft Model of Renal Medullary Carcinoma Demonstrates Therapeutic Efficacy of Sunitinib. Front. Oncol. 2021, 11, 648097. [Google Scholar] [CrossRef] [PubMed]
  34. Golan, T.; Stossel, C.; Schvimer, M.; Atias, D.; Halperin, S.; Buzhor, E.; Raitses-Gurevich, M.; Cohen, K.; Pri-Chen, S.; Wilson, J.; et al. Pancreatic cancer ascites xenograft—An expeditious model mirroring advanced therapeutic resistant disease. Oncotarget 2017, 8, 40778–40790. [Google Scholar] [CrossRef] [Green Version]
  35. Hodgkinson, C.L.; Morrow, C.J.; Li, Y.; Metcalf, R.L.; Rothwell, D.G.; Trapani, F.; Polanski, R.; Burt, D.J.; Simpson, K.L.; Morris, K.; et al. Tumorigenicity and genetic profiling of circulating tumor cells in small-cell lung cancer. Nat. Med. 2014, 20, 897–903. [Google Scholar] [CrossRef]
  36. Cho, S.-Y.; Kang, W.; Han, J.Y.; Min, S.; Kang, J.; Lee, A.; Kwon, J.Y.; Lee, C.; Park, H. An Integrative Approach to Precision Cancer Medicine Using Patient-Derived Xenografts. Mol. Cells 2016, 39, 77–86. [Google Scholar] [CrossRef] [Green Version]
  37. Tentler, J.J.; Tan, A.C.; Weekes, C.D.; Jimeno, A.; Leong, S.; Pitts, T.M.; Arcaroli, J.J.; Messersmith, W.A.; Eckhardt, S.G. Pa-tient-derived tumour xenografts as models for oncology drug development. Nat. Rev. Clin. Oncol. 2012, 9, 338–350. [Google Scholar] [CrossRef] [Green Version]
  38. Fichtner, I.; Rolff, J.; Soong, R.; Hoffmann, J.; Hammer, S.; Sommer, A.; Becker, M.; Merk, J. Establishment of Patient-Derived Non-Small Cell Lung Cancer Xenografts as Models for the Identification of Predictive Biomarkers. Clin. Cancer Res. 2008, 14, 6456–6468. [Google Scholar] [CrossRef] [Green Version]
  39. Fleming, J.M.; Miller, T.C.; Meyer, M.J.; Ginsburg, E.; Vonderhaar, B.K. Local regulation of human breast xenograft models. J. Cell. Physiol. 2010, 224, 795–806. [Google Scholar] [CrossRef] [Green Version]
  40. Petrillo, L.A.; Wolf, D.M.; Kapoun, A.M.; Wang, N.J.; Barczak, A.; Xiao, Y.; Korkaya, H.; Baehner, F.; Lewicki, J.; Wicha, M.; et al. Xenografts faithfully recapitulate breast cancer-specific gene expression patterns of parent primary breast tumors. Breast Cancer Res. Treat. 2012, 135, 913–922. [Google Scholar] [CrossRef]
  41. Kim, M.P.; Evans, D.B.; Wang, H.; Abbruzzese, J.L.; Fleming, J.B.; Gallick, G.E. Generation of orthotopic and heterotopic human pancreatic cancer xenografts in immunodeficient mice. Nat. Protoc. 2009, 4, 1670–1680. [Google Scholar] [CrossRef] [PubMed]
  42. Guenot, D.; Guerin, E.; Aguillon-Romain, S.; Pencreach, E.; Schneider, A.; Neuville, A.; Chenard, M.-P.; Duluc, I.; du Manoir, S.; Brigand, C.; et al. Primary tumour genetic alterations and intra-tumoral heterogeneity are maintained in xenografts of human colon cancers showing chromosome instability. J. Pathol. 2006, 208, 643–652. [Google Scholar] [CrossRef] [PubMed]
  43. Seol, H.S.; Kang, H.; Lee, S.-I.; Kim, N.E.; Kim, T.I.; Chun, S.M.; Yu, C.S.; Suh, Y.-A.; Singh, S.R.; Chang, S.; et al. Development and characterization of a colon PDX model that reproduces drug responsiveness and the mutation profiles of its original tumor. Cancer Lett. 2014, 345, 56–64. [Google Scholar] [CrossRef] [PubMed]
  44. Corso, S.; Cargnelutti, M.; Durando, S.; Menegon, S.; Apicella, M.; Migliore, C.; Capeloa, T.; Ughetto, S.; Isella, C.; Medico, E.; et al. Rituximab Treatment Prevents Lymphoma Onset in Gastric Cancer Patient-Derived Xenografts. Neoplasia 2018, 20, 443–455. [Google Scholar] [CrossRef] [PubMed]
  45. Dobbin, Z.C.; Katre, A.A.; Steg, A.D.; Erickson, B.K.; Shah, M.M.; Alvarez, R.D.; Conner, M.G.; Schneider, D.; Chen, D.; Landen, C.N. Using heterogeneity of the patient-derived xenograft model to identify the chemoresistant population in ovarian cancer. Oncotarget 2014, 5, 8750–8764. [Google Scholar] [CrossRef] [Green Version]
  46. Gu, Q.; Zhang, B.; Sun, H.; Xu, Q.; Tan, Y.; Wang, G.; Luo, Q.; Xu, W.; Yang, S.; Li, J.; et al. Genomic characterization of a large panel of patient-derived hepatocellular carcinoma xenograft tumor models for preclinical development. Oncotarget 2015, 6, 20160–20176. [Google Scholar] [CrossRef] [Green Version]
  47. Beshiri, M.L.; Tice, C.M.; Tran, C.; Nguyen, H.M.; Sowalsky, A.G.; Agarwal, S.; Jansson, K.H.; Yang, Q.; McGowen, K.M.; Yin, J.; et al. A PDX/Organoid Biobank of Advanced Prostate Cancers Captures Genomic and Phenotypic Heterogeneity for Disease Modeling and Therapeutic Screening. Clin. Cancer Res. 2018, 24, 4332–4345. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Tsoli, M.; Shen, H.; Mayoh, C.; Franshaw, L.; Ehteda, A.; Upton, D.; Carvalho, D.; Vinci, M.; Meel, M.H.; van Vuurden, D.; et al. International experience in the development of patient-derived xenograft models of diffuse intrinsic pontine glioma. J. Neuro-Oncology 2018, 141, 253–263. [Google Scholar] [CrossRef]
  49. Xiao, M.; Rebecca, V.W.; Herlyn, M. A Melanoma Patient-Derived Xenograft Model. J. Vis. Exp. 2019, 147, e59508. [Google Scholar] [CrossRef]
  50. Li, H.; Wheeler, S.; Park, Y.; Ju, Z.; Thomas, S.M.; Fichera, M.; Egloff, A.M.; Lui, V.W.; Duvvuri, U.; Bauman, J.E.; et al. Pro-teomic Characterization of Head and Neck Cancer Patient-Derived Xenografts. Mol. Cancer Res. 2016, 14, 278–286. [Google Scholar] [CrossRef]
  51. Jung, J.; Lee, C.H.; Seol, H.S.; Choi, Y.S.; Kim, E.; Lee, E.J.; Rhee, J.-K.; Singh, S.R.; Jun, E.S.; Han, B.; et al. Generation and molecular characterization of pancreatic cancer patient-derived xenografts reveals their heterologous nature. Oncotarget 2016, 7, 62533–62546. [Google Scholar] [CrossRef] [Green Version]
  52. Williams, S.A.; Anderson, W.C.; Santaguida, M.T.; Dylla, S.J. Patient-derived xenografts, the cancer stem cell paradigm, and cancer pathobiology in the 21st century. Lab. Investig. 2013, 93, 970–982. [Google Scholar] [CrossRef] [Green Version]
  53. Gao, H.; Korn, J.M.; Ferretti, S.; Monahan, J.E.; Wang, Y.; Singh, M.; Zhang, C.; Schnell, C.; Yang, G.; Zhang, Y.; et al. High-throughput screening using patient-derived tumor xenografts to predict clinical trial drug response. Nat. Med. 2015, 21, 1318–1325. [Google Scholar] [CrossRef] [PubMed]
  54. Bertotti, A.; Migliardi, G.; Galimi, F.; Sassi, F.; Torti, D.; Isella, C.; Corà, D.; Di Nicolantonio, F.; Buscarino, M.; Petti, C.; et al. A Molecularly Annotated Platform of Patient-Derived Xenografts (“Xenopatients”) Identifies HER2 as an Effective Therapeutic Target in Cetuximab-Resistant Colorectal Cancer. Cancer Discov. 2011, 1, 508–523. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Leto, S.M.; Sassi, F.; Catalano, I.; Torri, V.; Migliardi, G.; Zanella, E.R.; Throsby, M.; Bertotti, A.; Trusolino, L. Sustained Inhi-bition of HER3 and EGFR Is Necessary to Induce Regression of HER2-Amplified Gastrointestinal Carcinomas. Clin. Cancer Res. 2015, 21, 5519–5531. [Google Scholar] [CrossRef] [Green Version]
  56. Rubio-Viqueira, B.; Jimeno, A.; Cusatis, G.; Zhang, X.; Iacobuzio-Donahue, C.; Karikari, C.; Shi, C.; Danenberg, K.; Danenberg, P.V.; Kuramochi, H.; et al. An In vivo Platform for Translational Drug Development in Pancreatic Cancer. Clin. Cancer Res. 2006, 12, 4652–4661. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Hidalgo, M.; Bruckheimer, E.; Rajeshkumar, N.; Garrido-Laguna, I.; De Oliveira, E.; Rubio-Viqueira, B.; Strawn, S.; Wick, M.J.; Martell, J.; Sidransky, D. A Pilot Clinical Study of Treatment Guided by Personalized Tumorgrafts in Patients with Advanced Cancer. Mol. Cancer Ther. 2011, 10, 1311–1316. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Stebbing, J.; Paz, K.; Schwartz, G.K.; Wexler, L.H.; Maki, R.G.; Pollock, R.E.; Morris, R.; Cohen, R.; Shankar, A.; Blackman, G.; et al. Patient-derived xenografts for individualized care in advanced sarcoma. Cancer 2014, 120, 2006–2015. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Damhofer, H.; Ebbing, E.A.; Steins, A.; Welling, L.; Tol, J.A.; Krishnadath, K.K.; Van Leusden, T.; Van De Vijver, M.J.; Besselink, M.G.; Busch, O.R.; et al. Establishment of patient-derived xenograft models and cell lines for malignancies of the upper gas-trointestinal tract. J. Transl. Med. 2015, 13, 115. [Google Scholar] [CrossRef] [Green Version]
  60. Sugaya, M.; Takenoyama, M.; Osaki, T.; Yasuda, M.; Nagashima, A.; Sugio, K.; Yasumoto, K. Establishment of 15 Cancer Cell Lines from Patients with Lung Cancer and the Potential Tools for Immunotherapy. Chest 2002, 122, 282–288. [Google Scholar] [CrossRef]
  61. Meijer, T.G.; Naipal, K.A.; Jager, A.; van Gent, D.C. Ex vivo tumor culture systems for functional drug testing and therapy response prediction. Futur. Sci. OA 2017, 3, FSO190. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Liu, X.; Ory, V.; Chapman, S.; Yuan, H.; Albanese, C.; Kallakury, B.; Timofeeva, O.A.; Nealon, C.; Dakic, A.; Simic, V.; et al. ROCK Inhibitor and Feeder Cells Induce the Conditional Reprogramming of Epithelial Cells. Am. J. Pathol. 2012, 180, 599–607. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Suprynowicz, F.A.; Upadhyay, G.; Krawczyk, E.; Kramer, S.C.; Hebert, J.D.; Liu, X.; Yuan, H.; Cheluvaraju, C.; Clapp, P.W.; Boucher, R.C., Jr.; et al. Conditionally reprogrammed cells represent a stem-like state of adult epithelial cells. Proc. Natl. Acad. Sci. USA 2012, 109, 20035–20040. [Google Scholar] [CrossRef] [Green Version]
  64. Liu, X.; Krawczyk, E.; Suprynowicz, F.A.; Palechor-Ceron, N.; Yuan, H.; Dakic, A.; Simic, V.; Zheng, Y.-L.; Sripadhan, P.; Chen, C.; et al. Conditional reprogramming and long-term expansion of normal and tumor cells from human biospecimens. Nat. Protoc. 2017, 12, 439–451. [Google Scholar] [CrossRef] [PubMed]
  65. Chen, C.; Choudhury, S.; Wangsa, D.; Lescott, C.J.; Wilkins, D.J.; Sripadhan, P.; Liu, X.; Wangsa, D.; Ried, T.; Moskaluk, C.; et al. A multiplex preclinical model for adenoid cystic carcinoma of the salivary gland identifies regorafenib as a potential ther-apeutic drug. Sci. Rep. 2017, 7, 1–15. [Google Scholar] [CrossRef]
  66. Mahajan, A.S.; Sugita, B.; Duttargi, A.N.; Saenz, F.R.; Krawczyk, E.; McCutcheon, J.N.; Fonseca, A.S.; Kallakury, B.; Pohl-mann, P.; Gusev, Y.; et al. Genomic comparison of early-passage conditionally reprogrammed breast cancer cells to their cor-responding primary tumors. PLoS ONE 2017, 12, e0186190. [Google Scholar] [CrossRef] [Green Version]
  67. Timofeeva, O.A.; Palechor-Ceron, N.; Li, G.; Yuan, H.; Krawczyk, E.; Zhong, X.; Liu, G.; Upadhyay, G.; Dakic, A.; Yu, S.; et al. Conditionally reprogrammed normal and primary tumor prostate epithelial cells: A novel patient-derived cell model for studies of human prostate cancer. Oncotarget 2016, 8, 22741–22758. [Google Scholar] [CrossRef] [Green Version]
  68. Saeed, K.; Rahkama, V.; Eldfors, S.; Bychkov, D.; Mpindi, J.P.; Yadav, B.; Paavolainen, L.; Aittokallio, T.; Heckman, C.; Wennerberg, K.; et al. Comprehensive Drug Testing of Patient-derived Conditionally Reprogrammed Cells from Castra-tion-resistant Prostate Cancer. Eur. Urol. 2017, 71, 319–327. [Google Scholar] [CrossRef] [Green Version]
  69. Beglyarova, N.; Banina, E.; Zhou, Y.; Mukhamadeeva, R.; Andrianov, G.; Bobrov, E.; Lysenko, E.; Skobeleva, N.; Gabitova, L.; Restifo, D.; et al. Screening of Conditionally Reprogrammed Patient-Derived Carcinoma Cells Identifies ERCC3–MYC Inter-actions as a Target in Pancreatic Cancer. Clin. Cancer Res. 2016, 22, 6153–6163. [Google Scholar] [CrossRef] [Green Version]
  70. Li, Y.; Guo, D.; Zhang, Y.; Wang, L.; Sun, T.; Li, Z.; Zhang, X.; Wang, S.; Chen, Y.; Wu, A. Rapid screening for individualized chemotherapy optimization of colorectal cancer: A novel conditional reprogramming technology-based functional diagnostic assay. Transl. Oncol. 2020, 14, 100935. [Google Scholar] [CrossRef]
  71. Wu, M.; Hong, G.; Chen, Y.; Ye, L.; Zhang, K.; Cai, K.; Yang, H.; Long, X.; Gao, W.; Li, H. Personalized drug testing in a patient with non-small-cell lung cancer using cultured cancer cells from pleural effusion. J. Int. Med. Res. 2020, 48. [Google Scholar] [CrossRef] [PubMed]
  72. Correa, B.R.S.; Hu, J.; Penalva, L.; Schlegel, R.; Rimm, D.L.; Galante, P.A.F.; Agarwal, S. Patient-derived conditionally repro-grammed cells maintain intra-tumor genetic heterogeneity. Sci. Rep. 2018, 8, 1–7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Yuan, H.; Krawczyk, E.; Blancato, J.; Albanese, C.; Zhou, D.; Wang, N.; Paul, S.; Alkhilaiwi, F.; Palechor-Ceron, N.; Dakic, A.; et al. HPV positive neuroendocrine cervical cancer cells are dependent on Myc but not E6/E7 viral oncogenes. Sci. Rep. 2017, 7, 45617. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Alamri, A.M.; Kang, K.; Groeneveld, S.; Wang, W.; Zhong, X.; Kallakury, B.; Hennighausen, L.; Liu, X.; Furth, P.A. Primary cancer cell culture: Mammary-optimized vs conditional reprogramming. Endocrine-Related Cancer 2016, 23, 535–554. [Google Scholar] [CrossRef] [Green Version]
  75. Borodovsky, A.; McQuiston, T.J.; Stetson, D.; Ahmed, A.; Whitston, D.; Zhang, J.; Grondine, M.; Lawson, D.; Challberg, S.S.; Zinda, M.; et al. Generation of stable PDX derived cell lines using conditional reprogramming. Mol. Cancer 2017, 16, 177. [Google Scholar] [CrossRef] [Green Version]
  76. Mondal, A.M.; Ma, A.-H.; Li, G.; Krawczyk, E.; Yuan, R.; Lu, J.; Schlegel, R.; Stamatakis, L.; Kowalczyk, K.J.; Philips, G.K.; et al. Fidelity of a PDX-CR model for bladder cancer. Biochem. Biophys. Res. Commun. 2019, 517, 49–56. [Google Scholar] [CrossRef]
  77. Choudhary, S.; Ramasundaram, P.; Dziopa, E.; Mannion, C.; Kissin, Y.; Tricoli, L.; Albanese, C.; Lee, W.; Zilberberg, J. Human ex vivo 3D bone model recapitulates osteocyte response to metastatic prostate cancer. Sci. Rep. 2018, 8, 1–12. [Google Scholar] [CrossRef] [Green Version]
  78. Palechor-Ceron, N.; Krawczyk, E.; Dakic, A.; Simic, V.; Yuan, H.; Blancato, J.; Wang, W.; Hubbard, F.; Zheng, Y.-L.; Dan, H.; et al. Conditional Reprogramming for Patient-Derived Cancer Models and Next-Generation Living Biobanks. Cells 2019, 8, 1327. [Google Scholar] [CrossRef] [Green Version]
  79. Luo, Y.; Ju, L.; Wang, G.; Chen, C.; Wang, Y.; Chen, L.; Zhang, Y.; Xiao, Y.; Wang, X. Comprehensive genomic profiling of urothelial carcinoma cell lines reveals hidden research bias and caveats. Clin. Transl. Med. 2020, 10, 294–296. [Google Scholar] [CrossRef]
  80. Mimoto, R.; Yogosawa, S.; Saijo, H.; Fushimi, A.; Nogi, H.; Asakura, T.; Yoshida, K.; Takeyama, H. Clinical implications of drug-screening assay for recurrent metastatic hormone receptor-positive, human epidermal receptor 2-negative breast cancer using conditionally reprogrammed cells. Sci. Rep. 2019, 9, 1–8. [Google Scholar] [CrossRef]
  81. Alamri, A.M.; Liu, X.; Blancato, J.K.; Haddad, B.R.; Wang, W.; Zhong, X.; Choudhary, S.; Krawczyk, E.; Kallakury, B.V.; Davidson, B.J.; et al. Expanding primary cells from mucoepidermoid and other salivary gland neoplasms for genetic and chemosensitivity testing. Dis. Model. Mech. 2018, 11, dmm031716. [Google Scholar] [CrossRef] [Green Version]
  82. Kurihara, M. Clinical Experience with UFT in Japan. Adv. Exp. Med. Biol. 1993, 339, 243–251. [Google Scholar] [CrossRef] [PubMed]
  83. Tsushima, T.; Hironaka, S.; Boku, N.; Machida, N.; Yamazaki, K.; Yasui, H.; Taku, K.; Fukutomi, A.; Onozawa, Y. Safety and efficacy of S-1 monotherapy in elderly patients with advanced gastric cancer. Gastric Cancer 2010, 13, 245–250. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Wen, L.; You, C.; Lu, X.; Zhang, L. Phase II trial of concurrent chemoradiotherapy with S-1 versus weekly cisplatin for lo-coregionally advanced nasopharyngeal carcinoma. Mol. Clin. Oncol. 2015, 3, 687–691. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Sato, T.; Vries, R.G.; Snippert, H.J.; Van De Wetering, M.; Barker, N.; Stange, D.E.; Van Es, J.H.; Abo, A.; Kujala, P.; Peters, P.J.; et al. Single Lgr5 Stem Cells Build Crypt-Villus Structures in Vitro without a Mesenchymal Niche. Nature 2009, 459, 262–265. [Google Scholar] [CrossRef] [PubMed]
  86. Driehuis, E.; Kretzschmar, K.; Clevers, H. Establishment of patient-derived cancer organoids for drug-screening applications. Nat. Protoc. 2020, 15, 3380–3409, Erratum in Nat. Protoc. 2021, 16, 5739. [Google Scholar] [CrossRef]
  87. Drost, J.; Karthaus, W.R.; Gao, D.; Driehuis, E.; Sawyers, C.L.; Chen, Y.; Clevers, H. Organoid culture systems for prostate epithelial and cancer tissue. Nat. Protoc. 2016, 11, 347–358. [Google Scholar] [CrossRef] [Green Version]
  88. Kopper, O.; de Witte, C.J.; Lõhmussaar, K.; Valle-Inclan, J.E.; Hami, N.; Kester, L.; Balgobind, A.V.; Korving, J.; Proost, N.; Begthel, H.; et al. An organoid platform for ovarian cancer captures intra- and interpatient heterogeneity. Nat. Med. 2019, 25, 838–849. [Google Scholar] [CrossRef]
  89. Pauli, C.; Hopkins, B.D.; Prandi, D.; Shaw, R.; Fedrizzi, T.; Sboner, A.; Sailer, V.; Augello, M.; Puca, L.; Rosati, R.; et al. Per-sonalized in vitro and in vivo cancer models to guide precision medicine. Cancer Discov. 2017, 7, 462–477. [Google Scholar] [CrossRef] [Green Version]
  90. Lee, S.H.; Hu, W.; Matulay, J.T.; Silva, M.V.; Owczarek, T.B.; Kim, K.; Chua, C.W.; Barlow, L.M.J.; Kandoth, C.; Williams, A.B.; et al. Tumor Evolution and Drug Response in Patient-Derived Organoid Models of Bladder Cancer. Cell 2018, 173, 515–528.e17. [Google Scholar] [CrossRef]
  91. Hill, S.J.; Decker, B.; Roberts, E.A.; Horowitz, N.S.; Muto, M.G.; Worley, M.J.; Feltmate, C.M.; Nucci, M.R.; Swisher, E.M.; Nguyen, H.; et al. Prediction of DNA Repair Inhibitor Response in Short-Term Patient-Derived Ovarian Cancer Organoids. Cancer Discov. 2018, 8, 1404–1421. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Vlachogiannis, G.; Hedayat, S.; Vatsiou, A.; Jamin, Y.; Fernández-Mateos, J.; Khan, K.; Lampis, A.; Eason, K.; Huntingford, I.; Burke, R.; et al. Patient-derived organoids model treatment response of metastatic gastrointestinal cancers. Science 2018, 359, 920–926. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Tiriac, H.; Belleau, P.; Engle, D.D.; Plenker, D.; Deschenes, A.; Somerville, T.D.D.; Froeling, F.E.M.; Burkhart, R.A.; Denroche, R.E.; Jang, G.H.; et al. Organoid Profiling Identifies Common Responders to Chemotherapy in Pancreatic Cancer. Cancer Discov. 2018, 8, 1112–1129. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Yao, Y.; Xu, X.; Yang, L.; Zhu, J.; Wan, J.; Shen, L.; Xia, F.; Fu, G.; Deng, Y.; Pan, M.; et al. Patient-Derived Organoids Predict Chemoradiation Responses of Locally Advanced Rectal Cancer. Cell Stem Cell 2020, 26, 17–26.E16. [Google Scholar] [CrossRef]
  95. Friedman, A.A.; Letai, A.; Fisher, D.E.; Flaherty, K.T. Precision medicine for cancer with next-generation functional diagnostics. Nat. Rev. Cancer 2015, 15, 747–756. [Google Scholar] [CrossRef] [Green Version]
  96. Zhang, F.; Wang, W.; Long, Y.; Liu, H.; Cheng, J.; Guo, L.; Li, R.; Meng, C.; Yu, S.; Zhao, Q.; et al. Characterization of drug responses of mini patient-derived xenografts in mice for predicting cancer patient clinical therapeutic response. Cancer Commun. 2018, 38, 60–72. [Google Scholar] [CrossRef] [Green Version]
  97. Zhan, M.; Yang, R.-M.; Wang, H.; He, M.; Chen, W.; Xu, S.-W.; Yang, L.-H.; Liu, Q.; Long, M.-M.; Wang, J. Guided chemo-therapy based on patient-derived mini-xenograft models improves survival of gallbladder carcinoma patients. Cancer Commun. 2018, 38, 48–49. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Zhao, P.; Chen, H.; Wen, D.; Mou, S.; Zhang, F.; Zheng, S. Personalized treatment based on mini patient-derived xenografts and WES/RNA sequencing in a patient with metastatic duodenal adenocarcinoma. Cancer Commun. 2018, 38, 54–57. [Google Scholar] [CrossRef] [Green Version]
  99. Huang, Y.; Xu, J.; Li, K.; Wang, J.; Dai, Y.; Kang, Y. A Novel, Personalized Drug-Screening System for Platinum-Resistant Ovarian Cancer Patients: A Preliminary Clinical Report. Cancer Manag. Res. 2021, 13, 2849–2867. [Google Scholar] [CrossRef]
  100. Wang, X.; Sun, Y.; Xu, Y.; Wen, D.; An, N.; Leng, X.; Fu, G.; Lu, S.; Chen, Z. Mini-patient-derived xenograft assay based on microfluidic technology promises to be an effective tool for screening individualized chemotherapy regimens for advanced non-small cell lung cancer. Cell Biol. Int. 2021, 45, 1887–1896. [Google Scholar] [CrossRef]
  101. Li, C.; Sun, Y.-D.; Yu, G.-Y.; Cui, J.-R.; Lou, Z.; Zhang, H.; Huang, Y.; Bai, C.-G.; Deng, L.-L.; Liu, P.; et al. Integrated Omics of Metastatic Colorectal Cancer. Cancer Cell 2020, 38, 734–747.e9. [Google Scholar] [CrossRef] [PubMed]
  102. Yang, L.; Yuan, Z.; Zhang, Y.; Cui, Z.; Li, Y.; Hou, J.; Liu, X.; Liu, Z.; Shi, R.; Tian, Q.; et al. MiniPDX-guided postoperative anticancer treatment can effectively prolong the survival of patients with hepatocellular carcinoma. Cancer Chemother. Pharmacol. 2020, 87, 125–134. [Google Scholar] [CrossRef] [PubMed]
  103. Wang, J.; Sun, T.; Meng, Z.; Wang, L.; Li, M.; Chen, J.; Qin, T.; Yu, J.; Zhang, M.; Bie, Z.; et al. XPO1 inhibition synergizes with PARP1 inhibition in small cell lung cancer by targeting nuclear transport of FOXO3a. Cancer Lett. 2021, 503, 197–212. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Schematic representation of CR cells, organoid, MiniPDX and PDX. For CR cells, organoids, and MiniPDX tumor tissues are diced and digested enzymatically to collect the primary suspension cells. (A). Tumor cells are co-cultured with feeder cells (irradiated or mitomycin C-treated mouse fibroblasts) and CR medium added ROCK inhibitor Y-27632. (B). Patient-derived organoids are established by embedment into a 3D matrix and specific medium to grow into 3D organotypic structures. (C). PDX is established by subcutaneous or orthotopic implantation of surgical or biopsy tissue chunks into immune-deficient mice. (D). Primary cells are filled into OncoVee® capsules (LIDE Biotech Co., Ltd., Shanghai, China) and implanted subcutaneously via a small skin incision. The implanted capsules are removed after 7 days, and tumor cell proliferation is evaluated.
Figure 1. Schematic representation of CR cells, organoid, MiniPDX and PDX. For CR cells, organoids, and MiniPDX tumor tissues are diced and digested enzymatically to collect the primary suspension cells. (A). Tumor cells are co-cultured with feeder cells (irradiated or mitomycin C-treated mouse fibroblasts) and CR medium added ROCK inhibitor Y-27632. (B). Patient-derived organoids are established by embedment into a 3D matrix and specific medium to grow into 3D organotypic structures. (C). PDX is established by subcutaneous or orthotopic implantation of surgical or biopsy tissue chunks into immune-deficient mice. (D). Primary cells are filled into OncoVee® capsules (LIDE Biotech Co., Ltd., Shanghai, China) and implanted subcutaneously via a small skin incision. The implanted capsules are removed after 7 days, and tumor cell proliferation is evaluated.
Cells 11 03868 g001
Table 1. Characteristics of cancer models. The advantages and limitations of the cancer models mentioned are listed in Table 1, including features of biology, methodology, and application.
Table 1. Characteristics of cancer models. The advantages and limitations of the cancer models mentioned are listed in Table 1, including features of biology, methodology, and application.
FeaturePDXMiniPDXOrganoidConditional ReprogrammingCDXGEMMSyngeneic Model
Success rate of initiationLowHighMediumMediumHighHighHigh
HumanizationYesYesYesYesYesNoNo
Initial sample sourceFresh clinical specimensFresh clinical specimensFresh clinical specimensFresh clinical specimensHuman cancer cell line-Mouse cancer cell line
Medium-dependencyNoNoYesYesNoNoNo
Administration approaches SystemicallySystemicallyThrough mediumThrough mediumSystemi-callySystem-icallySystemica-lly
Numbers of animals neededHighLow--HighHighHigh
Facility requirementsHighHighLowLowHighMediumMedium
CostHighMediumLowLowMedium to highHighMedium to high
High-throughput drug screeningNoNoYesYesNoNoNo
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Long, Y.; Xie, B.; Shen, H.C.; Wen, D. Translation Potential and Challenges of In Vitro and Murine Models in Cancer Clinic. Cells 2022, 11, 3868. https://doi.org/10.3390/cells11233868

AMA Style

Long Y, Xie B, Shen HC, Wen D. Translation Potential and Challenges of In Vitro and Murine Models in Cancer Clinic. Cells. 2022; 11(23):3868. https://doi.org/10.3390/cells11233868

Chicago/Turabian Style

Long, Yuan, Bin Xie, Hong C. Shen, and Danyi Wen. 2022. "Translation Potential and Challenges of In Vitro and Murine Models in Cancer Clinic" Cells 11, no. 23: 3868. https://doi.org/10.3390/cells11233868

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop