1
|
Yildiz M, Romano A, Xanthoulea S. Murine Xenograft Models as Preclinical Tools in Endometrial Cancer Research. Cancers (Basel) 2024; 16:3994. [PMID: 39682182 DOI: 10.3390/cancers16233994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 11/22/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
Murine xenograft models are valuable and increasingly used preclinical tools in cancer research to understand disease pathogenesis and guide treatment options. The aim of this narrative review is to summarize the studies that employed mouse xenograft models, using cell lines, patient-derived tumors, or organoids, in endometrial cancer (EC) research, detailing their methodology and main findings. We identified 27 articles reporting on heterotopic EC xenografts, including subcutaneous, subrenal capsule, intraperitoneal, and retro-orbital models, and 18 articles using orthotopic xenografts. Subcutaneous xenografts generated using either cell lines or patient tumors have been widely used; however, their low engraftment rates and the inability to recapitulate main clinical features such as metastases limit their translational value. Subrenal capsule models showed improved engraftment rates compared to subcutaneous models, but tumors exhibited slower and constrained tumor growth. Orthotopic models are technically more challenging to generate and monitor, but tumor growth occurs in a relevant microenvironment and EC ortho-xenografts exhibit high engraftment rates and metastases to clinically relevant sites. Cell line-based xenograft (CDX) models are attractive tools because they are convenient, easy to use, and amenable to genetic modifications, making them suitable for proof-of-concept approaches and large-scale studies. EC xenografts developed from patient tumors (PDTXs) are more labor/cost-intensive for their establishment but can capture the genetic and molecular heterogeneity within and across histologic subtypes and can inform personalized patient treatment. EC organoid-based xenograft (PDOX) models combine the advantages of both CDXs and PDTXs since they are more time- and cost-effective, faithfully maintain tumor characteristics and therapeutic responses, and can be genetically modified. Despite substantial progress in EC management, there are still several unmet needs. Efficient targeted treatments are currently indicated only for a small subgroup of patients, while women with recurrent or advanced-stage EC have very few therapeutic options and their prognosis remains unfavorable. Novel (targeted) drugs, combinational regimens and tools to predict the real drug response in patients are urgently needed. Xenograft models are expected to inform about disease mechanisms and to help identify novel therapeutic options and suitable target patients.
Collapse
Affiliation(s)
- Merve Yildiz
- GROW-Research Institute for Oncology & Reproduction, Maastricht University, 6229 ER Maastricht, The Netherlands
- Department of Obstetrics and Gynaecology, Maastricht University Medical Centre, 6229 HX Maastricht, The Netherlands
| | - Andrea Romano
- GROW-Research Institute for Oncology & Reproduction, Maastricht University, 6229 ER Maastricht, The Netherlands
- Department of Obstetrics and Gynaecology, Maastricht University Medical Centre, 6229 HX Maastricht, The Netherlands
| | - Sofia Xanthoulea
- GROW-Research Institute for Oncology & Reproduction, Maastricht University, 6229 ER Maastricht, The Netherlands
- Department of Obstetrics and Gynaecology, Maastricht University Medical Centre, 6229 HX Maastricht, The Netherlands
| |
Collapse
|
2
|
Vaklavas C, Matsen CB, Chu Z, Boucher KM, Scherer SD, Pathi S, Beck A, Brownson KE, Buys SS, Chittoria N, D'Astous E, Gulbahce HE, Henry NL, Kimani S, Porretta J, Rosenthal R, Ward J, Wei M, Welm BE, Welm AL. TOWARDS Study: Patient-Derived Xenograft Engraftment Predicts Poor Survival in Patients With Newly Diagnosed Triple-Negative Breast Cancer. JCO Precis Oncol 2024; 8:e2300724. [PMID: 39074345 PMCID: PMC11371112 DOI: 10.1200/po.23.00724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 05/04/2024] [Accepted: 05/28/2024] [Indexed: 07/31/2024] Open
Abstract
PURPOSE Assessing risk of recurrence for nonmetastatic triple-negative breast cancer (TNBC) is a key determinant of therapeutic strategy. The best predictor of recurrence risk is failure to achieve a pathologic complete response after preoperative chemotherapy, but it imperfectly correlates with the definitive end points of relapse-free and overall survival (OS). The inability to accurately predict recurrence has led to increasingly toxic treatment regimens for patients with early-stage TNBC. Better assays for recurrence risk are needed to tailor aggressive therapy for patients who need it and avoid overtreatment and unnecessary toxicity for those at low risk. The purpose of this study was to determine if patient-derived xenograft (PDX) engraftment of newly diagnosed breast tumors can serve as an accurate predictor of recurrence and death from breast cancer. METHODS This study was a blinded noninterventional trial comprising 80 patients with newly diagnosed, nonmetastatic, estrogen receptor (ER)-negative or ER-low breast cancer. RESULTS PDX engraftment was strongly associated with relapse in 1 year: 8 of 18 (44.4%) patients whose tumors engrafted relapsed versus 1 of 62 (1.6%) patients whose tumors did not engraft (P < .0001). Patients whose tumors engrafted had a hazard ratio (HR) for relapse of 17.5. HRs for OS and breast cancer-specific survival in PDX+ patients were 21.1 and 39.5, respectively. CONCLUSION We report that the ability of a tumor to engraft as a PDX predicts early recurrence by serving as a functional readout of aggressiveness and prospectively identifies the most devastating tumors. This provides new opportunity to develop surrogate assays, such as biomarkers of engraftment, which will extend the clinical feasibility of this finding.
Collapse
Affiliation(s)
- Christos Vaklavas
- Department of Internal Medicine, Division of Oncology, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | - Cindy B Matsen
- Department of Surgery, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | - Zhengtao Chu
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | - Kenneth M Boucher
- Department of Internal Medicine, Division of Epidemiology, Salt Lake City, UT
| | - Sandra D Scherer
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | - Satya Pathi
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | - Anna Beck
- Department of Internal Medicine, Division of Oncology, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | - Kirstyn E Brownson
- Department of Surgery, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | - Saundra S Buys
- Department of Internal Medicine, Division of Oncology, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | - Namita Chittoria
- Department of Internal Medicine, Division of Oncology, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | - Elyse D'Astous
- Huntsman Cancer Institute Clinical Trials Office, Salt Lake City, UT
| | - H Evin Gulbahce
- Department of Pathology, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | - N Lynn Henry
- Department of Internal Medicine, Division of Oncology, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
- Current Address: Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| | - Stephen Kimani
- Department of Internal Medicine, Division of Oncology, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | - Jane Porretta
- Department of Surgery, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | - Regina Rosenthal
- Department of Surgery, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | - John Ward
- Department of Internal Medicine, Division of Oncology, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | - Mei Wei
- Department of Internal Medicine, Division of Oncology, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | - Bryan E Welm
- Department of Surgery, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | - Alana L Welm
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| |
Collapse
|
3
|
Ueda S, Tanaka T, Hirosuna K, Miyamoto S, Murakami H, Nishie R, Tsuchihashi H, Toji A, Morita N, Hashida S, Daimon A, Terada S, Maruoka H, Kogata Y, Taniguchi K, Komura K, Ohmichi M. Consistency between Primary Uterine Corpus Malignancies and Their Corresponding Patient-Derived Xenograft Models. Int J Mol Sci 2024; 25:1486. [PMID: 38338763 PMCID: PMC10855170 DOI: 10.3390/ijms25031486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 01/23/2024] [Accepted: 01/23/2024] [Indexed: 02/12/2024] Open
Abstract
Patient-derived xenograft (PDX) models retain the characteristics of tumors and are useful tools for personalized therapy and translational research. In this study, we aimed to establish PDX models for uterine corpus malignancies (UC-PDX) and analyze their similarities. Tissue fragments obtained from 92 patients with uterine corpus malignancies were transplanted subcutaneously into immunodeficient mice. Histological and immunohistochemical analyses were performed to compare tumors of patients with PDX tumors. DNA and RNA sequencing were performed to validate the genetic profile. Furthermore, the RNA in extracellular vesicles (EVs) extracted from primary and PDX tumors was analyzed. Among the 92 cases, 52 UC-PDX models were established, with a success rate of 56.5%. The success rate depended on tumor histology and staging. The pathological and immunohistochemical features of primary and PDX tumors were similar. DNA sequencing revealed similarities in gene mutations between the primary and PDX tumors. RNA sequencing showed similarities in gene expressions between primary and PDX tumors. Furthermore, the RNA profiles of the EVs obtained from primary and PDX tumors were similar. As UC-PDX retained the pathological and immunohistochemical features and gene profiles of primary tumors, they may provide a platform for developing personalized medicine and translational research.
Collapse
Affiliation(s)
- Shoko Ueda
- Department of Obstetrics and Gynecology, Educational Foundation of Osaka Medical and Pharmaceutical University, 2-7 Daigakumachi, Takatsuki 569-8686, Osaka, Japan; (S.U.); (S.M.); (H.M.); (R.N.); (H.T.); (A.T.); (N.M.); (S.H.); (A.D.); (S.T.); (H.M.); (Y.K.); (M.O.)
| | - Tomohito Tanaka
- Department of Obstetrics and Gynecology, Educational Foundation of Osaka Medical and Pharmaceutical University, 2-7 Daigakumachi, Takatsuki 569-8686, Osaka, Japan; (S.U.); (S.M.); (H.M.); (R.N.); (H.T.); (A.T.); (N.M.); (S.H.); (A.D.); (S.T.); (H.M.); (Y.K.); (M.O.)
- Center for Medical Research & Development, Division of Translational Research, Osaka Medical and Pharmaceutical University, 2-7 Daigakumachi, Takatsuki 569-8686, Osaka, Japan; (K.T.); (K.K.)
| | - Kensuke Hirosuna
- Department of Regenerative Science, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikatachou, Kita-ku, Okayama 700-8558, Okayama, Japan;
| | - Shunsuke Miyamoto
- Department of Obstetrics and Gynecology, Educational Foundation of Osaka Medical and Pharmaceutical University, 2-7 Daigakumachi, Takatsuki 569-8686, Osaka, Japan; (S.U.); (S.M.); (H.M.); (R.N.); (H.T.); (A.T.); (N.M.); (S.H.); (A.D.); (S.T.); (H.M.); (Y.K.); (M.O.)
- Center for Medical Research & Development, Division of Translational Research, Osaka Medical and Pharmaceutical University, 2-7 Daigakumachi, Takatsuki 569-8686, Osaka, Japan; (K.T.); (K.K.)
| | - Hikaru Murakami
- Department of Obstetrics and Gynecology, Educational Foundation of Osaka Medical and Pharmaceutical University, 2-7 Daigakumachi, Takatsuki 569-8686, Osaka, Japan; (S.U.); (S.M.); (H.M.); (R.N.); (H.T.); (A.T.); (N.M.); (S.H.); (A.D.); (S.T.); (H.M.); (Y.K.); (M.O.)
| | - Ruri Nishie
- Department of Obstetrics and Gynecology, Educational Foundation of Osaka Medical and Pharmaceutical University, 2-7 Daigakumachi, Takatsuki 569-8686, Osaka, Japan; (S.U.); (S.M.); (H.M.); (R.N.); (H.T.); (A.T.); (N.M.); (S.H.); (A.D.); (S.T.); (H.M.); (Y.K.); (M.O.)
| | - Hiromitsu Tsuchihashi
- Department of Obstetrics and Gynecology, Educational Foundation of Osaka Medical and Pharmaceutical University, 2-7 Daigakumachi, Takatsuki 569-8686, Osaka, Japan; (S.U.); (S.M.); (H.M.); (R.N.); (H.T.); (A.T.); (N.M.); (S.H.); (A.D.); (S.T.); (H.M.); (Y.K.); (M.O.)
| | - Akihiko Toji
- Department of Obstetrics and Gynecology, Educational Foundation of Osaka Medical and Pharmaceutical University, 2-7 Daigakumachi, Takatsuki 569-8686, Osaka, Japan; (S.U.); (S.M.); (H.M.); (R.N.); (H.T.); (A.T.); (N.M.); (S.H.); (A.D.); (S.T.); (H.M.); (Y.K.); (M.O.)
| | - Natsuko Morita
- Department of Obstetrics and Gynecology, Educational Foundation of Osaka Medical and Pharmaceutical University, 2-7 Daigakumachi, Takatsuki 569-8686, Osaka, Japan; (S.U.); (S.M.); (H.M.); (R.N.); (H.T.); (A.T.); (N.M.); (S.H.); (A.D.); (S.T.); (H.M.); (Y.K.); (M.O.)
| | - Sousuke Hashida
- Department of Obstetrics and Gynecology, Educational Foundation of Osaka Medical and Pharmaceutical University, 2-7 Daigakumachi, Takatsuki 569-8686, Osaka, Japan; (S.U.); (S.M.); (H.M.); (R.N.); (H.T.); (A.T.); (N.M.); (S.H.); (A.D.); (S.T.); (H.M.); (Y.K.); (M.O.)
| | - Atsushi Daimon
- Department of Obstetrics and Gynecology, Educational Foundation of Osaka Medical and Pharmaceutical University, 2-7 Daigakumachi, Takatsuki 569-8686, Osaka, Japan; (S.U.); (S.M.); (H.M.); (R.N.); (H.T.); (A.T.); (N.M.); (S.H.); (A.D.); (S.T.); (H.M.); (Y.K.); (M.O.)
| | - Shinichi Terada
- Department of Obstetrics and Gynecology, Educational Foundation of Osaka Medical and Pharmaceutical University, 2-7 Daigakumachi, Takatsuki 569-8686, Osaka, Japan; (S.U.); (S.M.); (H.M.); (R.N.); (H.T.); (A.T.); (N.M.); (S.H.); (A.D.); (S.T.); (H.M.); (Y.K.); (M.O.)
| | - Hiroshi Maruoka
- Department of Obstetrics and Gynecology, Educational Foundation of Osaka Medical and Pharmaceutical University, 2-7 Daigakumachi, Takatsuki 569-8686, Osaka, Japan; (S.U.); (S.M.); (H.M.); (R.N.); (H.T.); (A.T.); (N.M.); (S.H.); (A.D.); (S.T.); (H.M.); (Y.K.); (M.O.)
| | - Yuhei Kogata
- Department of Obstetrics and Gynecology, Educational Foundation of Osaka Medical and Pharmaceutical University, 2-7 Daigakumachi, Takatsuki 569-8686, Osaka, Japan; (S.U.); (S.M.); (H.M.); (R.N.); (H.T.); (A.T.); (N.M.); (S.H.); (A.D.); (S.T.); (H.M.); (Y.K.); (M.O.)
| | - Kohei Taniguchi
- Center for Medical Research & Development, Division of Translational Research, Osaka Medical and Pharmaceutical University, 2-7 Daigakumachi, Takatsuki 569-8686, Osaka, Japan; (K.T.); (K.K.)
| | - Kazumasa Komura
- Center for Medical Research & Development, Division of Translational Research, Osaka Medical and Pharmaceutical University, 2-7 Daigakumachi, Takatsuki 569-8686, Osaka, Japan; (K.T.); (K.K.)
| | - Masahide Ohmichi
- Department of Obstetrics and Gynecology, Educational Foundation of Osaka Medical and Pharmaceutical University, 2-7 Daigakumachi, Takatsuki 569-8686, Osaka, Japan; (S.U.); (S.M.); (H.M.); (R.N.); (H.T.); (A.T.); (N.M.); (S.H.); (A.D.); (S.T.); (H.M.); (Y.K.); (M.O.)
| |
Collapse
|
4
|
Liang F, Xu H, Cheng H, Zhao Y, Zhang J. Patient-derived tumor models: a suitable tool for preclinical studies on esophageal cancer. Cancer Gene Ther 2023; 30:1443-1455. [PMID: 37537209 DOI: 10.1038/s41417-023-00652-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 07/13/2023] [Accepted: 07/26/2023] [Indexed: 08/05/2023]
Abstract
Esophageal cancer (EC) is the tenth most common cancer worldwide and has high morbidity and mortality. Its main subtypes include esophageal squamous cell carcinoma and esophageal adenocarcinoma, which are usually diagnosed during their advanced stages. The biological defects and inability of preclinical models to summarize completely the etiology of multiple factors, the complexity of the tumor microenvironment, and the genetic heterogeneity of tumors severely limit the clinical treatment of EC. Patient-derived models of EC not only retain the tissue structure, cell morphology, and differentiation characteristics of the original tumor, they also retain tumor heterogeneity. Therefore, compared with other preclinical models, they can better predict the efficacy of candidate drugs, explore novel biomarkers, combine with clinical trials, and effectively improve patient prognosis. This review discusses the methods and animals used to establish patient-derived models and genetically engineered mouse models, especially patient-derived xenograft models. It also discusses their advantages, applications, and limitations as preclinical experimental research tools to provide an important reference for the precise personalized treatment of EC and improve the prognosis of patients.
Collapse
Affiliation(s)
- Fan Liang
- Institutes of Health Central Plains, Xinxiang Medical University, Xinxiang, 453003, China
| | - Hongyan Xu
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China
| | - Hongwei Cheng
- Institutes of Health Central Plains, Xinxiang Medical University, Xinxiang, 453003, China
| | - Yabo Zhao
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China
| | - Junhe Zhang
- Institutes of Health Central Plains, Xinxiang Medical University, Xinxiang, 453003, China.
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China.
| |
Collapse
|
5
|
Liu L, Wu M, Huang A, Gao C, Yang Y, Liu H, Jiang H, Yu L, Huang Y, Wang H. Establishment of a high-fidelity patient-derived xenograft model for cervical cancer enables the evaluation of patient's response to conventional and novel therapies. J Transl Med 2023; 21:611. [PMID: 37689699 PMCID: PMC10492358 DOI: 10.1186/s12967-023-04444-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 08/16/2023] [Indexed: 09/11/2023] Open
Abstract
BACKGROUND Recurrent or metastatic cervical cancer (r/m CC) often has poor prognosis owing to its limited treatment options. The development of novel therapeutic strategies has been hindered by the lack of preclinical models that accurately reflect the biological and genomic heterogeneity of cervical cancer (CC). Herein, we aimed to establish a large patient-derived xenograft (PDX) biobank for CC, evaluate the consistency of the biologic indicators between PDX and primary tumor tissues of patients, and explore its utility for assessing patient's response to conventional and novel therapies. METHODS Sixty-nine fresh CC tumor tissues were implanted directly into immunodeficient mice to establish PDX models. The concordance of the PDX models with their corresponding primary tumors (PTs) was compared based on the clinical pathological features, protein biomarker levels, and genomic features through hematoxylin & eosin staining, immunohistochemistry, and whole exome sequencing, respectively. Moreover, the clinical information of CC patients, RNA transcriptome and immune phenotyping of primary tumors were integrated to identify the potential parameters that could affect the success of xenograft engraftment. Subsequently, PDX model was evaluated for its capacity to mirror patient's response to chemotherapy. Finally, PDX model and PDX-derived organoid (PDXO) were utilized to evaluate the therapeutic efficacy of neratinib and adoptive cell therapy (ACT) combination strategy for CC patients with human epidermal growth factor receptor 2 (HER2) mutation. RESULTS We established a PDX biobank for CC with a success rate of 63.8% (44/69). The primary features of established PDX tumors, including clinicopathological features, the expression levels of protein biomarkers including Ki67, α-smooth muscle actin, and p16, and genomics, were highly consistent with their PTs. Furthermore, xenograft engraftment was likely influenced by the primary tumor size, the presence of follicular helper T cells and the expression of cell adhesion-related genes in primary tumor tissue. The CC derived PDX models were capable of recapitulating the patient's response to chemotherapy. In a PDX model, a novel therapeutic strategy, the combination of ACT and neratinib, was shown to effectively inhibit the growth of PDX tumors derived from CC patients with HER2-mutation. CONCLUSIONS We established by far the largest PDX biobank with a high engraftment rate for CC that preserves the histopathological and genetic characteristics of patient's biopsy samples, recapitulates patient's response to conventional therapy, and is capable of evaluating the efficacy of novel therapeutic modalities for CC.
Collapse
Affiliation(s)
- Liting Liu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Min Wu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Anni Huang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chun Gao
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yifan Yang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hong Liu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Han Jiang
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Long Yu
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yafei Huang
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Hui Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Zhejiang Provincial Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| |
Collapse
|
6
|
Berube LL, Nickel KOP, Iida M, Ramisetty S, Kulkarni P, Salgia R, Wheeler DL, Kimple RJ. Radiation Sensitivity: The Rise of Predictive Patient-Derived Cancer Models. Semin Radiat Oncol 2023; 33:279-286. [PMID: 37331782 PMCID: PMC10287034 DOI: 10.1016/j.semradonc.2023.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Patient-derived cancer models have been used for decades to improve our understanding of cancer and test anticancer treatments. Advances in radiation delivery have made these models more attractive for studying radiation sensitizers and understanding an individual patient's radiation sensitivity. Advances in the use of patient-derived cancer models lead to a more clinically relevant outcome, although many questions remain regarding the optimal use of patient-derived xenografts and patient-derived spheroid cultures. The use of patient-derived cancer models as personalized predictive avatars through mouse and zebrafish models is discussed, and the advantages and disadvantages of patient-derived spheroids are reviewed. In addition, the use of large repositories of patient-derived models to develop predictive algorithms to guide treatment selection is discussed. Finally, we review methods for establishing patient-derived models and identify key factors that influence their use as both avatars and models of cancer biology.
Collapse
Affiliation(s)
- Liliana L Berube
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI; Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Kwang-Ok P Nickel
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Mari Iida
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Sravani Ramisetty
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, CA
| | - Prakash Kulkarni
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, CA
| | - Ravi Salgia
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, CA
| | - Deric L Wheeler
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI; University of Wisconsin Carbone Cancer Center, Madison, WI
| | - Randall J Kimple
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI; Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI; University of Wisconsin Carbone Cancer Center, Madison, WI.
| |
Collapse
|
7
|
Liu Y, Wu W, Cai C, Zhang H, Shen H, Han Y. Patient-derived xenograft models in cancer therapy: technologies and applications. Signal Transduct Target Ther 2023; 8:160. [PMID: 37045827 PMCID: PMC10097874 DOI: 10.1038/s41392-023-01419-2] [Citation(s) in RCA: 130] [Impact Index Per Article: 65.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 03/21/2023] [Indexed: 04/14/2023] Open
Abstract
Patient-derived xenograft (PDX) models, in which tumor tissues from patients are implanted into immunocompromised or humanized mice, have shown superiority in recapitulating the characteristics of cancer, such as the spatial structure of cancer and the intratumor heterogeneity of cancer. Moreover, PDX models retain the genomic features of patients across different stages, subtypes, and diversified treatment backgrounds. Optimized PDX engraftment procedures and modern technologies such as multi-omics and deep learning have enabled a more comprehensive depiction of the PDX molecular landscape and boosted the utilization of PDX models. These irreplaceable advantages make PDX models an ideal choice in cancer treatment studies, such as preclinical trials of novel drugs, validating novel drug combinations, screening drug-sensitive patients, and exploring drug resistance mechanisms. In this review, we gave an overview of the history of PDX models and the process of PDX model establishment. Subsequently, the review presents the strengths and weaknesses of PDX models and highlights the integration of novel technologies in PDX model research. Finally, we delineated the broad application of PDX models in chemotherapy, targeted therapy, immunotherapy, and other novel therapies.
Collapse
Affiliation(s)
- Yihan Liu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
| | - Wantao Wu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
| | - Changjing Cai
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
| | - Hao Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Hong Shen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China.
| | - Ying Han
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China.
| |
Collapse
|
8
|
Devis‐Jauregui L, Vidal A, Plata‐Peña L, Santacana M, García‐Mulero S, Bonifaci N, Noguera‐Delgado E, Ruiz N, Gil M, Dorca E, Llobet FJ, Coll‐Iglesias L, Gassner K, Martinez‐Iniesta M, Rodriguez‐Barrueco R, Barahona M, Marti L, Viñals F, Ponce J, Sanz‐Pamplona R, Piulats JM, Vivancos A, Matias‐Guiu X, Villanueva A, Llobet‐Navas D. Generation and Integrated Analysis of Advanced Patient-Derived Orthoxenograft Models (PDOX) for the Rational Assessment of Targeted Therapies in Endometrial Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 10:e2204211. [PMID: 36373729 PMCID: PMC9811454 DOI: 10.1002/advs.202204211] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 10/18/2022] [Indexed: 05/19/2023]
Abstract
Clinical management of endometrial cancer (EC) is handicapped by the limited availability of second line treatments and bona fide molecular biomarkers to predict recurrence. These limitations have hampered the treatment of these patients, whose survival rates have not improved over the last four decades. The advent of coordinated studies such as The Cancer Genome Atlas Uterine Corpus Endometrial Carcinoma (TCGA_UCEC) has partially solved this issue, but the lack of proper experimental systems still represents a bottleneck that precludes translational studies from successful clinical testing in EC patients. Within this context, the first study reporting the generation of a collection of endometrioid-EC-patient-derived orthoxenograft (PDOX) mouse models is presented that is believed to overcome these experimental constraints and pave the way toward state-of-the-art precision medicine in EC. The collection of primary tumors and derived PDOXs is characterized through an integrative approach based on transcriptomics, mutational profiles, and morphological analysis; and it is demonstrated that EC tumors engrafted in the mouse uterus retain the main molecular and morphological features from analogous tumor donors. Finally, the molecular properties of these tumors are harnessed to assess the therapeutic potential of trastuzumab, a human epidermal growth factor receptor 2 (HER2) inhibitor with growing interest in EC, using patient-derived organotypic multicellular tumor spheroids and in vivo experiments.
Collapse
|
9
|
Chen H, Zheng M, Zhang W, Long Y, Xu Y, Yuan M. Research Status of Mouse Models for Non-Small-Cell Lung Cancer (NSCLC) and Antitumor Therapy of Traditional Chinese Medicine (TCM) in Mouse Models. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2022; 2022:6404853. [PMID: 36185084 PMCID: PMC9519343 DOI: 10.1155/2022/6404853] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 08/31/2022] [Indexed: 11/25/2022]
Abstract
Non-small-cell lung cancer (NSCLC) is known as one of the most lethal cancers, causing more than 1 million deaths annually worldwide. Therefore, the development of novel therapeutic drugs for NSCLC has become an urgent need. Herein, various mouse models provide great convenience not only for researchers but also for the development of antitumor drug. Meanwhile, TCM, as a valuable and largely untapped resource pool for modern medicine, provides research resources for the treatment of various diseases. Until now, cell-derived xenograft (CDX) model, patient-derived xenograft (PDX) model, syngeneic model, orthotopic model, humanized mouse model (HIS), and genetically engineered mouse models (GEMMs) have been reported in TCM evaluation. This review shows the role and current status of kinds of mouse models in antitumor research and summarizes the application progress of TCM including extracts, formulas, and isolated single molecules for NSCLC therapy in various mouse models; more importantly, it provides a theoretical exploration of what kind of mouse models is ideal for TCM efficacy evaluation in future. However, there are still huge challenges and limitations in the development of mouse models specifically for the TCM research, and none of the available models are perfectly matching the characteristics of TCM, which suppress the tumor growth through various mechanisms, especially by regulating immune function. Nevertheless, with fully functional immune system existing in syngeneic model and humanized mouse model (HIS), it is still suggested that these two models are more suitable for development of TCM especially for TCM extracts or formulas. Moreover, continued efforts are needed to generate more reliable mouse models to test TCM formulas in future research.
Collapse
Affiliation(s)
- Hongkui Chen
- Shanghai Lidebiotech Co. Ltd., Shanghai 201203, China
| | - Min Zheng
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Wenhui Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai 201203, China
| | - Yuan Long
- Shanghai Lidebiotech Co. Ltd., Shanghai 201203, China
| | - Yu Xu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai 201203, China
| | - Man Yuan
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai 201203, China
| |
Collapse
|
10
|
Qin T, Fan J, Lu F, Zhang L, Liu C, Xiong Q, Zhao Y, Chen G, Sun C. Harnessing preclinical models for the interrogation of ovarian cancer. J Exp Clin Cancer Res 2022; 41:277. [PMID: 36114548 PMCID: PMC9479310 DOI: 10.1186/s13046-022-02486-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Accepted: 09/05/2022] [Indexed: 12/24/2022] Open
Abstract
Ovarian cancer (OC) is a heterogeneous malignancy with various etiology, histopathology, and biological feature. Despite accumulating understanding of OC in the post-genomic era, the preclinical knowledge still undergoes limited translation from bench to beside, and the prognosis of ovarian cancer has remained dismal over the past 30 years. Henceforth, reliable preclinical model systems are warranted to bridge the gap between laboratory experiments and clinical practice. In this review, we discuss the status quo of ovarian cancer preclinical models which includes conventional cell line models, patient-derived xenografts (PDXs), patient-derived organoids (PDOs), patient-derived explants (PDEs), and genetically engineered mouse models (GEMMs). Each model has its own strengths and drawbacks. We focus on the potentials and challenges of using these valuable tools, either alone or in combination, to interrogate critical issues with OC.
Collapse
|
11
|
Tanaka T, Nishie R, Ueda S, Miyamoto S, Hashida S, Konishi H, Terada S, Kogata Y, Sasaki H, Tsunetoh S, Taniguchi K, Komura K, Ohmichi M. Endometrial Cancer Patient-Derived Xenograft Models: A Systematic Review. J Clin Med 2022; 11:2606. [PMID: 35566732 PMCID: PMC9100787 DOI: 10.3390/jcm11092606] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 04/29/2022] [Accepted: 05/04/2022] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Because patient-derived xenograft (PDX) models resemble the original tumors, they can be used as platforms to find target agents for precision medicine and to study characteristics of tumor biology such as clonal evolution and microenvironment interactions. The aim of this review was to identify articles on endometrial cancer PDXs (EC-PDXs) and verify the methodology and outcomes. METHODS We used PubMed to research and identify articles on EC-PDX. The data were analyzed descriptively. RESULTS Post literature review, eight studies were selected for the systematic review. Eighty-five EC-PDXs were established from 173 patients with EC, with a total success rate of 49.1%. A 1-10 mm3 fragment was usually implanted. Fresh-fragment implantation had higher success rates than using overnight-stored or frozen fragments. Primary tumors were successfully established with subcutaneous implantation, but metastasis rarely occurred; orthotopic implantation via minced tumor cell injection was better for metastatic models. The success rate did not correspond to immunodeficiency grades, and PDXs using nude mice reduced costs. The tumor growth period ranged from 2 weeks to 13 months. Similar characteristics were observed between primary tumors and PDXs, including pathological findings, gene mutations, and gene expression. CONCLUSION EC-PDXs are promising tools for translational research because they closely resemble the features of tumors in patients and retain molecular and histological features of the disease.
Collapse
Affiliation(s)
- Tomohito Tanaka
- Department of Obstetrics and Gynecology, Educational Foundation of Osaka Medical and Pharmaceutical University, 2-7 Daigakumachi, Takatsuki, Osaka 569-8686, Japan; (R.N.); (S.U.); (S.M.); (S.H.); (H.K.); (S.T.); (Y.K.); (H.S.); (S.T.); (M.O.)
- Translational Research Program, Educational Foundation of Osaka Medical and Pharmaceutical University, 2-7 Daigakumachi, Takatsuki, Osaka 569-8686, Japan; (K.T.); (K.K.)
| | - Ruri Nishie
- Department of Obstetrics and Gynecology, Educational Foundation of Osaka Medical and Pharmaceutical University, 2-7 Daigakumachi, Takatsuki, Osaka 569-8686, Japan; (R.N.); (S.U.); (S.M.); (S.H.); (H.K.); (S.T.); (Y.K.); (H.S.); (S.T.); (M.O.)
| | - Shoko Ueda
- Department of Obstetrics and Gynecology, Educational Foundation of Osaka Medical and Pharmaceutical University, 2-7 Daigakumachi, Takatsuki, Osaka 569-8686, Japan; (R.N.); (S.U.); (S.M.); (S.H.); (H.K.); (S.T.); (Y.K.); (H.S.); (S.T.); (M.O.)
| | - Shunsuke Miyamoto
- Department of Obstetrics and Gynecology, Educational Foundation of Osaka Medical and Pharmaceutical University, 2-7 Daigakumachi, Takatsuki, Osaka 569-8686, Japan; (R.N.); (S.U.); (S.M.); (S.H.); (H.K.); (S.T.); (Y.K.); (H.S.); (S.T.); (M.O.)
| | - Sousuke Hashida
- Department of Obstetrics and Gynecology, Educational Foundation of Osaka Medical and Pharmaceutical University, 2-7 Daigakumachi, Takatsuki, Osaka 569-8686, Japan; (R.N.); (S.U.); (S.M.); (S.H.); (H.K.); (S.T.); (Y.K.); (H.S.); (S.T.); (M.O.)
| | - Hiromi Konishi
- Department of Obstetrics and Gynecology, Educational Foundation of Osaka Medical and Pharmaceutical University, 2-7 Daigakumachi, Takatsuki, Osaka 569-8686, Japan; (R.N.); (S.U.); (S.M.); (S.H.); (H.K.); (S.T.); (Y.K.); (H.S.); (S.T.); (M.O.)
| | - Shinichi Terada
- Department of Obstetrics and Gynecology, Educational Foundation of Osaka Medical and Pharmaceutical University, 2-7 Daigakumachi, Takatsuki, Osaka 569-8686, Japan; (R.N.); (S.U.); (S.M.); (S.H.); (H.K.); (S.T.); (Y.K.); (H.S.); (S.T.); (M.O.)
| | - Yuhei Kogata
- Department of Obstetrics and Gynecology, Educational Foundation of Osaka Medical and Pharmaceutical University, 2-7 Daigakumachi, Takatsuki, Osaka 569-8686, Japan; (R.N.); (S.U.); (S.M.); (S.H.); (H.K.); (S.T.); (Y.K.); (H.S.); (S.T.); (M.O.)
| | - Hiroshi Sasaki
- Department of Obstetrics and Gynecology, Educational Foundation of Osaka Medical and Pharmaceutical University, 2-7 Daigakumachi, Takatsuki, Osaka 569-8686, Japan; (R.N.); (S.U.); (S.M.); (S.H.); (H.K.); (S.T.); (Y.K.); (H.S.); (S.T.); (M.O.)
| | - Satoshi Tsunetoh
- Department of Obstetrics and Gynecology, Educational Foundation of Osaka Medical and Pharmaceutical University, 2-7 Daigakumachi, Takatsuki, Osaka 569-8686, Japan; (R.N.); (S.U.); (S.M.); (S.H.); (H.K.); (S.T.); (Y.K.); (H.S.); (S.T.); (M.O.)
| | - Kohei Taniguchi
- Translational Research Program, Educational Foundation of Osaka Medical and Pharmaceutical University, 2-7 Daigakumachi, Takatsuki, Osaka 569-8686, Japan; (K.T.); (K.K.)
| | - Kazumasa Komura
- Translational Research Program, Educational Foundation of Osaka Medical and Pharmaceutical University, 2-7 Daigakumachi, Takatsuki, Osaka 569-8686, Japan; (K.T.); (K.K.)
| | - Masahide Ohmichi
- Department of Obstetrics and Gynecology, Educational Foundation of Osaka Medical and Pharmaceutical University, 2-7 Daigakumachi, Takatsuki, Osaka 569-8686, Japan; (R.N.); (S.U.); (S.M.); (S.H.); (H.K.); (S.T.); (Y.K.); (H.S.); (S.T.); (M.O.)
| |
Collapse
|
12
|
Shapiro DD, Virumbrales-Muñoz M, Beebe DJ, Abel EJ. Models of Renal Cell Carcinoma Used to Investigate Molecular Mechanisms and Develop New Therapeutics. Front Oncol 2022; 12:871252. [PMID: 35463327 PMCID: PMC9022005 DOI: 10.3389/fonc.2022.871252] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 03/10/2022] [Indexed: 12/24/2022] Open
Abstract
Modeling renal cell carcinoma is critical to investigating tumor biology and therapeutic mechanisms. Multiple systems have been developed to represent critical components of the tumor and its surrounding microenvironment. Prominent in vitro models include traditional cell cultures, 3D organoid models, and microphysiological devices. In vivo models consist of murine patient derived xenografts or genetically engineered mice. Each system has unique advantages as well as limitations and researchers must thoroughly understand each model to properly investigate research questions. This review addresses common model systems for renal cell carcinoma and critically evaluates their performance and ability to measure tumor characteristics.
Collapse
Affiliation(s)
- Daniel D Shapiro
- Department of Urology, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States.,Division of Urology, William S. Middleton Memorial Veterans Hospital, Madison, WI, United States
| | - Maria Virumbrales-Muñoz
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States.,Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - David J Beebe
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States.,Department of Biomedical Engineering, University of Wisconsin - Madison, Madison, WI, United States
| | - E Jason Abel
- Department of Urology, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| |
Collapse
|