351
|
Jang SY, Bae HI, Lee IK, Park HK, Cho CM. Successful Xenograft of Endoscopic Ultrasound-Guided Fine-Needle Aspiration Specimen from Human Extrahepatic Cholangiocarcinoma into an Immunodeficient Mouse. Gut Liver 2016; 9:805-8. [PMID: 26087785 PMCID: PMC4625712 DOI: 10.5009/gnl14279] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Patient-derived tumor xenograft is the transfer of primary human tumors directly into an immunodeficient mouse. Patient-derived tumor xenograft plays an important role in the development and evaluation of new chemotherapeutic agents. We succeeded in generating a patient-derived tumor xenograft of a biliary tumor obtained by endoscopic ultrasound-guided fine-needle aspiration from a patient who had an inoperable extrahepatic cholangiocarcinoma. This patient-derived tumor xenograft will be a promising tool for individualized cancer therapy and can be used in developing new chemotherapeutic agents for the treatment of biliary cancer in the future.
Collapse
Affiliation(s)
- Se Young Jang
- Department of Internal Medicine, Kyungpook National University School of Medicine, Daegu, Korea
| | - Han Ik Bae
- Department of Pathology, Kyungpook National University School of Medicine, Daegu, Korea
| | - In Kyu Lee
- Department of Internal Medicine, Kyungpook National University School of Medicine, Daegu, Korea.,BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University Hospital, Daegu, Korea.,Leading-Edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease (DMRC), Kyungpook National University Hospital, Daegu, Korea
| | - Hwan Ki Park
- Leading-Edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease (DMRC), Kyungpook National University Hospital, Daegu, Korea
| | - Chang-Min Cho
- Department of Internal Medicine, Kyungpook National University School of Medicine, Daegu, Korea
| |
Collapse
|
352
|
Bondarenko G, Ugolkov A, Rohan S, Kulesza P, Dubrovskyi O, Gursel D, Mathews J, O'Halloran TV, Wei JJ, Mazar AP. Patient-Derived Tumor Xenografts Are Susceptible to Formation of Human Lymphocytic Tumors. Neoplasia 2016; 17:735-741. [PMID: 26476081 PMCID: PMC4611072 DOI: 10.1016/j.neo.2015.09.004] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Revised: 09/08/2015] [Accepted: 09/15/2015] [Indexed: 01/01/2023] Open
Abstract
Patient-derived xenograft (PDX) tumor models have emerged as a new approach to evaluate the effects of cancer drugs on patients’ personalized tumor grafts enabling to select the best treatment for the cancer patient and providing a new tool for oncology drug developers. Here, we report that human tumors engrafted in immunodeficient mice are susceptible to formation of B-and T-cell PDX tumors. We xenografted human primary and metastatic tumor samples into immunodeficient mice and found that a fraction of PDX tumors generated from patients’ samples of breast, colon, pancreatic, bladder and renal cancer were histologically similar to lymphocytic neoplasms. Moreover, we found that the first passage of breast and pancreatic cancer PDX tumors after initial transplantation of the tumor pieces from the same human tumor graft could grow as a lymphocytic tumor in one mouse and as an adenocarcinoma in another mouse. Whereas subcutaneous PDX tumors resembling human adenocarcinoma histology were slow growing and non-metastatic, we found that subcutaneous PDX lymphocytic tumors were fast growing and formed large metastatic lesions in mouse lymph nodes, liver, lungs, and spleen. PDX lymphocytic tumors were comprised of B-cells which were Epstein-Barr virus positive and expressed CD45 and CD20. Because B-cells are typically present in malignant solid tumors, formation of B-cell tumor may evolve in a wide range of PDX tumor models. Although PDX tumor models show great promise in the development of personalized therapy for cancer patients, our results suggest that confidence in any given PDX tumor model requires careful screening of lymphocytic markers.
Collapse
Affiliation(s)
- Gennadiy Bondarenko
- Center for Developmental Therapeutics, Robert H. Lurie Comprehensive Cancer Center, Northwestern University, 2170 Campus Drive, Evanston, IL, USA; Chemistry of Life Processes Institute, Northwestern University, 2170 Campus Drive, Evanston, IL, USA
| | - Andrey Ugolkov
- Center for Developmental Therapeutics, Robert H. Lurie Comprehensive Cancer Center, Northwestern University, 2170 Campus Drive, Evanston, IL, USA; Chemistry of Life Processes Institute, Northwestern University, 2170 Campus Drive, Evanston, IL, USA
| | - Stephen Rohan
- Department of Pathology, Feinberg School of Medicine, Northwestern University, 303 East Chicago Avenue, Chicago, 60611, IL, USA; Pathology Core Facility, Robert H. Lurie Comprehensive Cancer Center, Northwestern University, 710 North Fairbanks Court, Chicago, IL, USA
| | - Piotr Kulesza
- Department of Pathology, Feinberg School of Medicine, Northwestern University, 303 East Chicago Avenue, Chicago, 60611, IL, USA; Pathology Core Facility, Robert H. Lurie Comprehensive Cancer Center, Northwestern University, 710 North Fairbanks Court, Chicago, IL, USA
| | - Oleksii Dubrovskyi
- Center for Developmental Therapeutics, Robert H. Lurie Comprehensive Cancer Center, Northwestern University, 2170 Campus Drive, Evanston, IL, USA; Chemistry of Life Processes Institute, Northwestern University, 2170 Campus Drive, Evanston, IL, USA
| | - Demirkan Gursel
- Pathology Core Facility, Robert H. Lurie Comprehensive Cancer Center, Northwestern University, 710 North Fairbanks Court, Chicago, IL, USA
| | - Jeremy Mathews
- Pathology Core Facility, Robert H. Lurie Comprehensive Cancer Center, Northwestern University, 710 North Fairbanks Court, Chicago, IL, USA
| | - Thomas V O'Halloran
- Chemistry of Life Processes Institute, Northwestern University, 2170 Campus Drive, Evanston, IL, USA
| | - Jian J Wei
- Department of Pathology, Feinberg School of Medicine, Northwestern University, 303 East Chicago Avenue, Chicago, 60611, IL, USA; Pathology Core Facility, Robert H. Lurie Comprehensive Cancer Center, Northwestern University, 710 North Fairbanks Court, Chicago, IL, USA
| | - Andrew P Mazar
- Center for Developmental Therapeutics, Robert H. Lurie Comprehensive Cancer Center, Northwestern University, 2170 Campus Drive, Evanston, IL, USA; Department of Pharmacology, Feinberg School of Medicine, Northwestern University, 320 East Superior Street, Chicago, 60611, IL, USA; Chemistry of Life Processes Institute, Northwestern University, 2170 Campus Drive, Evanston, IL, USA.
| |
Collapse
|
353
|
Agorku DJ, Tomiuk S, Klingner K, Wild S, Rüberg S, Zatrieb L, Bosio A, Schueler J, Hardt O. Depletion of Mouse Cells from Human Tumor Xenografts Significantly Improves Downstream Analysis of Target Cells. J Vis Exp 2016:54259. [PMID: 27501218 PMCID: PMC5091706 DOI: 10.3791/54259] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
The use of in vitro cell line models for cancer research has been a useful tool. However, it has been shown that these models fail to reliably mimic patient tumors in different assays(1). Human tumor xenografts represent the gold standard with respect to tumor biology, drug discovery, and metastasis research (2-4). Tumor xenografts can be derived from different types of material like tumor cell lines, tumor tissue from primary patient tumors(4) or serially transplanted tumors. When propagated in vivo, xenografted tissue is infiltrated and vascularized by cells of mouse origin. Multiple factors such as the tumor entity, the origin of xenografted material, growth rate and region of transplantation influence the composition and the amount of mouse cells present in tumor xenografts. However, even when these factors are kept constant, the degree of mouse cell contamination is highly variable. Contaminating mouse cells significantly impair downstream analyses of human tumor xenografts. As mouse fibroblasts show high plating efficacies and proliferation rates, they tend to overgrow cultures of human tumor cells, especially slowly proliferating subpopulations. Mouse cell derived DNA, mRNA, and protein components can bias downstream gene expression analysis, next-generation sequencing, as well as proteome analysis (5). To overcome these limitations, we have developed a fast and easy method to isolate untouched human tumor cells from xenografted tumor tissue. This procedure is based on the comprehensive depletion of cells of mouse origin by combining automated tissue dissociation with the benchtop tissue dissociator and magnetic cell sorting. Here, we demonstrate that human target cells can be can be obtained with purities higher than 96% within less than 20 min independent of the tumor type.
Collapse
|
354
|
Abstract
Ras proteins are considered as the founding members of a large superfamily of small GTPases that control fundamental cellular functions. Mutationally activated RAS genes were discovered in human cancer cells more than 3 decades ago, but intensive efforts on Ras structure, biochemistry, function and signaling continue even now. Because mutant Ras proteins are inherently difficult to inhibit and have yet been therapeutically conquered, it was designated as “the Everest of oncogenes” in the cancer genome landscape, further promoting a “renaissance” in RAS research. Different paths to directly or indirectly targeting mutant Ras signaling are currently under investigation in the hope of finding an efficacious regimen. Inhibitors directly binding to KRASG12C to block its downstream signaling have been revealed, supporting the notion of Ras' druggability. An alternative indirect approach by targeting synthetic lethal interactors of mutant RAS is underway. We recently employed a synthetic lethal drug screen plus a combinatorial strategy using a panel of clinical agents and discovered that KRAS-mutant cancers were fragile to the combined inhibition of polo-like kinase 1 (Plk1) and RhoA/Rho kinase (ROCK). The combined regimen of BI-2536 (a Plk1 inhibitor) and fasudil (a ROCK inhibitor) promoted a significant inhibition of patient-derived lung cancer xenografts and prolonged the survival of LSL-KRASG12D mice. In this commentary, we will summarize the state-of-the art for the direction of synthetic lethality, and also speculate on the future development of this approach.
Collapse
Affiliation(s)
- Xiufeng Pang
- a Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University , Shanghai , China
| | - Mingyao Liu
- a Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University , Shanghai , China.,b Institute of Biosciences and Technology , Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center , Houston , TX , USA
| |
Collapse
|
355
|
Lee JY, Kim SY, Park C, Kim NKD, Jang J, Park K, Yi JH, Hong M, Ahn T, Rath O, Schueler J, Kim ST, Do IG, Lee S, Park SH, Ji YI, Kim D, Park JO, Park YS, Kang WK, Kim KM, Park WY, Lim HY, Lee J. Patient-derived cell models as preclinical tools for genome-directed targeted therapy. Oncotarget 2016; 6:25619-30. [PMID: 26296973 PMCID: PMC4694854 DOI: 10.18632/oncotarget.4627] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2015] [Accepted: 07/03/2015] [Indexed: 12/26/2022] Open
Abstract
Background In this study, we established patient-derived tumor cell (PDC) models using tissues collected from patients with metastatic cancer and assessed whether these models could be used as a tool for genome-based cancer treatment. Methods PDCs were isolated and cultured from malignant effusions including ascites and pleural fluid. Pathological examination, immunohistochemical analysis, and genomic profiling were performed to compare the histological and genomic features of primary tumors, PDCs. An exploratory gene expression profiling assay was performed to further characterize PDCs. Results From January 2012 to May 2013, 176 samples from patients with metastatic cancer were collected. PDC models were successfully established in 130 (73.6%) samples. The median time from specimen collection to passage 1 (P1) was 3 weeks (range, 0.5–4 weeks), while that from P1 to P2 was 2.5 weeks (range, 0.5–5 weeks). Sixteen paired samples of genomic alterations were highly concordant between each primary tumor and progeny PDCs, with an average variant allele frequency (VAF) correlation of 0.878. We compared genomic profiles of the primary tumor (P0), P1 cells, P2 cells, and patient-derived xenografts (PDXs) derived from P2 cells and found that three samples (P0, P1, and P2 cells) were highly correlated (0.99–1.00). Moreover, PDXs showed more than 100 variants, with correlations of only 0.6–0.8 for the other samples. Drug responses of PDCs were reflective of the clinical response to targeted agents in selected patient PDC lines. Conclusion(s) Our results provided evidence that our PDC model was a promising model for preclinical experiments and closely resembled the patient tumor genome and clinical response.
Collapse
Affiliation(s)
- Ji Yun Lee
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Sun Young Kim
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Charny Park
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Nayoung K D Kim
- Samsung Genome Institute, Samsung Medical Center, Seoul, Korea
| | - Jiryeon Jang
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Kyunghee Park
- Samsung Genome Institute, Samsung Medical Center, Seoul, Korea
| | - Jun Ho Yi
- Division of Hematology-Oncology, Department of Medicine, Hanyang University Hospital, Seoul, Korea
| | - Mineui Hong
- Innovative Cancer Medicine Institute, Samsung Cancer Center, Samsung Medical Center, Seoul, Korea.,Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Taejin Ahn
- Samsung Genome Institute, Samsung Medical Center, Seoul, Korea
| | | | | | - Seung Tae Kim
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - In-Gu Do
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Sujin Lee
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Se Hoon Park
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Yong Ick Ji
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Dukwhan Kim
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Joon Oh Park
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.,Innovative Cancer Medicine Institute, Samsung Cancer Center, Samsung Medical Center, Seoul, Korea
| | - Young Suk Park
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Won Ki Kang
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Kyoung-Mee Kim
- Innovative Cancer Medicine Institute, Samsung Cancer Center, Samsung Medical Center, Seoul, Korea.,Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Woong-Yang Park
- Samsung Genome Institute, Samsung Medical Center, Seoul, Korea.,Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Ho Yeong Lim
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jeeyun Lee
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.,Innovative Cancer Medicine Institute, Samsung Cancer Center, Samsung Medical Center, Seoul, Korea
| |
Collapse
|
356
|
Acetylation at lysine 71 inactivates superoxide dismutase 1 and sensitizes cancer cells to genotoxic agents. Oncotarget 2016; 6:20578-91. [PMID: 26008972 PMCID: PMC4653027 DOI: 10.18632/oncotarget.3987] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 04/10/2015] [Indexed: 01/13/2023] Open
Abstract
Cancer cells are characterized by a high dependency on antioxidant enzymes to cope with the elevated rates of reactive oxygen species (ROS). Impairing antioxidant capacity in cancer cells disturbs the ROS homeostasis and exposes cancer cells to massive oxidative stress. In this study, we have discovered that superoxide dismutase 1 (SOD1), a major player in maintaining the cellular redox status, was acetylated at lysine 71. This acetylation, which was primarily deacetylated by Sirtuin 1 (SIRT1), suppressed the enzymatic activity of SOD1 via disrupting its association with copper chaperone for SOD1 (CCS). More importantly, genotoxic agents, such as camptothecin (CPT), induced SOD1 acetylation by disrupting its binding with SIRT1. CPT-induced SOD1 acetylation was stimulated by its provoked ROS, suggesting a positive feedback loop, in which ROS per se impairs the antioxidative defence of cancer cells and reinforces oxidative stress stimulated by anticancer agents. The intrinsic abundance of SOD1 acetylation varied among cancer cells, and high level of SOD1 acetylation was correlated with elevated sensitivity to CPT. Together, our findings gained mechanistic insights into how cytotoxic agents fine tune the intracellular ROS homeostasis to strengthen their anticancer effects, and suggested SOD1 acetylation as a candidate biomarker for predicting response to CPT-based chemotherapy.
Collapse
|
357
|
Guller A, Grebenyuk P, Shekhter A, Zvyagin A, Deyev SM. Bioreactor-Based Tumor Tissue Engineering. Acta Naturae 2016; 8:44-58. [PMID: 27795843 PMCID: PMC5081698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Indexed: 11/16/2022] Open
Abstract
This review focuses on modeling of cancer tumors using tissue engineering technology. Tumor tissue engineering (TTE) is a new method of three-dimensional (3D) simulation of malignant neoplasms. Design and development of complex tissue engineering constructs (TECs) that include cancer cells, cell-bearing scaffolds acting as the extracellular matrix, and other components of the tumor microenvironment is at the core of this approach. Although TECs can be transplanted into laboratory animals, the specific aim of TTE is the most realistic reproduction and long-term maintenance of the simulated tumor properties in vitro for cancer biology research and for the development of new methods of diagnosis and treatment of malignant neoplasms. Successful implementation of this challenging idea depends on bioreactor technology, which will enable optimization of culture conditions and control of tumor TECs development. In this review, we analyze the most popular bioreactor types in TTE and the emerging applications.
Collapse
Affiliation(s)
- A.E. Guller
- Macquarie University, Sydney, 2109, New South Wales, Australia
- ARC Centre of Excellence for Nanoscale BioPhotonics, Macquarie University, Sydney 2109, New South Wales, Australia
- Sechenov First Moscow State Medical University, Institute for Regenerative Medicine, 8, Trubetskaya Str., Moscow, 119992, Russia
- Lobachevsky Nizhniy Novgorod State University, 23, Gagarina Ave., Nizhniy Novgorod, 603950, Russia
| | | | - A.B. Shekhter
- Sechenov First Moscow State Medical University, Institute for Regenerative Medicine, 8, Trubetskaya Str., Moscow, 119992, Russia
| | - A.V. Zvyagin
- Macquarie University, Sydney, 2109, New South Wales, Australia
- ARC Centre of Excellence for Nanoscale BioPhotonics, Macquarie University, Sydney 2109, New South Wales, Australia
- Sechenov First Moscow State Medical University, Institute for Regenerative Medicine, 8, Trubetskaya Str., Moscow, 119992, Russia
- Lobachevsky Nizhniy Novgorod State University, 23, Gagarina Ave., Nizhniy Novgorod, 603950, Russia
| | - S. M. Deyev
- Lobachevsky Nizhniy Novgorod State University, 23, Gagarina Ave., Nizhniy Novgorod, 603950, Russia
- Institute of Bioorganic Chemistry, 16/10, Miklukho-Maklaya Str., Moscow, 117871, Russia
- National Research Tomsk Polytechnic University, 30, Lenina Ave., Tomsk, 634050, Russia
| |
Collapse
|
358
|
Lane AN, Higashi RM, Fan TWM. Preclinical models for interrogating drug action in human cancers using Stable Isotope Resolved Metabolomics (SIRM). Metabolomics 2016; 12:118. [PMID: 27489532 PMCID: PMC4968890 DOI: 10.1007/s11306-016-1065-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
AIMS In this review we compare the advantages and disadvantages of different model biological systems for determining the metabolic functions of cells in complex environments, how they may change in different disease states, and respond to therapeutic interventions. BACKGROUND All preclinical drug-testing models have advantages and drawbacks. We compare and contrast established cell, organoid and animal models with ex vivo organ or tissue culture and in vivo human experiments in the context of metabolic readout of drug efficacy. As metabolism reports directly on the biochemical state of cells and tissues, it can be very sensitive to drugs and/or other environmental changes. This is especially so when metabolic activities are probed by stable isotope tracing methods, which can also provide detailed mechanistic information on drug action. We have developed and been applying Stable Isotope-Resolved Metabolomics (SIRM) to examine metabolic reprogramming of human lung cancer cells in monoculture, in mouse xenograft/explant models, and in lung cancer patients in situ (Lane et al. 2011; T. W. Fan et al. 2011; T. W-M. Fan et al. 2012; T. W. Fan et al. 2012; Xie et al. 2014b; Ren et al. 2014a; Sellers et al. 2015b). We are able to determine the influence of the tumor microenvironment using these models. We have now extended the range of models to fresh human tissue slices, similar to those originally described by O. Warburg (Warburg 1923), which retain the native tissue architecture and heterogeneity with a paired benign versus cancer design under defined cell culture conditions. This platform offers an unprecedented human tissue model for preclinical studies on metabolic reprogramming of human cancer cells in their tissue context, and response to drug treatment (Xie et al. 2014a). As the microenvironment of the target human tissue is retained and individual patient's response to drugs is obtained, this platform promises to transcend current limitations of drug selection for clinical trials or treatments. CONCLUSIONS AND FUTURE WORK Development of ex vivo human tissue and animal models with humanized organs including bone marrow and liver show considerable promise for analyzing drug responses that are more relevant to humans. Similarly using stable isotope tracer methods with these improved models in advanced stages of the drug development pipeline, in conjunction with tissue biopsy is expected significantly to reduce the high failure rate of experimental drugs in Phase II and III clinical trials.
Collapse
Affiliation(s)
- Andrew N Lane
- Center for Environmental and Systems Biochemistry, University of Kentucky
| | - Richard M Higashi
- Center for Environmental and Systems Biochemistry, University of Kentucky
| | - Teresa W-M Fan
- Center for Environmental and Systems Biochemistry, University of Kentucky
| |
Collapse
|
359
|
Coleman CN, Higgins GS, Brown JM, Baumann M, Kirsch DG, Willers H, Prasanna PGS, Dewhirst MW, Bernhard EJ, Ahmed MM. Improving the Predictive Value of Preclinical Studies in Support of Radiotherapy Clinical Trials. Clin Cancer Res 2016; 22:3138-47. [PMID: 27154913 PMCID: PMC4930691 DOI: 10.1158/1078-0432.ccr-16-0069] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 03/21/2016] [Indexed: 01/16/2023]
Abstract
There is an urgent need to improve reproducibility and translatability of preclinical data to fully exploit opportunities for molecular therapeutics involving radiation and radiochemotherapy. For in vitro research, the clonogenic assay remains the current state-of-the-art of preclinical assays, whereas newer moderate and high-throughput assays offer the potential for rapid initial screening. Studies of radiation response modification by molecularly targeted agents can be improved using more physiologic 3D culture models. Elucidating effects on the cancer stem cells (CSC, and CSC-like) and developing biomarkers for defining targets and measuring responses are also important. In vivo studies are necessary to confirm in vitro findings, further define mechanism of action, and address immunomodulation and treatment-induced modification of the microenvironment. Newer in vivo models include genetically engineered and patient-derived xenograft mouse models and spontaneously occurring cancers in domesticated animals. Selection of appropriate endpoints is important for in vivo studies; for example, regrowth delay measures bulk tumor killing, whereas local tumor control assesses effects on CSCs. The reliability of individual assays requires standardization of procedures and cross-laboratory validation. Radiation modifiers must be tested as part of clinical standard of care, which includes radiochemotherapy for most tumors. Radiation models are compatible with but also differ from those used for drug screening. Furthermore, the mechanism of a drug as a chemotherapeutic agent may be different from its interaction with radiation and/or radiochemotherapy. This provides an opportunity to expand the use of molecular-targeted agents. Clin Cancer Res; 22(13); 3138-47. ©2016 AACR.
Collapse
Affiliation(s)
- C Norman Coleman
- Radiation Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute (NCI), NIH, Bethesda, Maryland.
| | - Geoff S Higgins
- Cancer Research UK/Medical Research Council, Oxford Institute for Radiation Oncology, University of Oxford, United Kingdom
| | - J Martin Brown
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California
| | - Michael Baumann
- OncoRay National Center for Radiation Research, Technische Universität Dresden/Helmholtz-Zenrtum Dresden-Rossendorf, Dresden, Germany and German Cancer Consortium, Dresden/German Cancer Research Center (DKFZ)
| | - David G Kirsch
- Departments of Radiation Oncology and Pharmacology and Cancer Biology, Duke University, Durham, North Carolina
| | - Henning Willers
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Pataje G S Prasanna
- Radiation Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute (NCI), NIH, Bethesda, Maryland
| | - Mark W Dewhirst
- Departments of Radiation Oncology, Pathology and Biomedical Engineering, Duke University, Durham, North Carolina
| | - Eric J Bernhard
- Radiation Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute (NCI), NIH, Bethesda, Maryland
| | - Mansoor M Ahmed
- Radiation Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute (NCI), NIH, Bethesda, Maryland
| |
Collapse
|
360
|
Chen YC, Ingram PN, Fouladdel S, McDermott SP, Azizi E, Wicha MS, Yoon E. High-Throughput Single-Cell Derived Sphere Formation for Cancer Stem-Like Cell Identification and Analysis. Sci Rep 2016; 6:27301. [PMID: 27292795 PMCID: PMC4904376 DOI: 10.1038/srep27301] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Accepted: 04/27/2016] [Indexed: 01/05/2023] Open
Abstract
Considerable evidence suggests that many malignancies are driven by a cellular compartment that displays stem cell properties. Cancer stem-like cells (CSCs) can be identified by expression of cell surface markers or enzymatic activity, but these methods are limited by phenotypic heterogeneity and plasticity of CSCs. An alternative phenotypic methodology based on in-vitro sphere formation has been developed, but it is typically labor-intensive and low-throughput. In this work, we present a 1,024-microchamber microfluidic platform for single-cell derived sphere formation. Utilizing a hydrodynamic capturing scheme, more than 70% of the microchambers capture only one cell, allowing for monitoring of sphere formation from heterogeneous cancer cell populations for identification of CSCs. Single-cell derived spheres can be retrieved and dissociated for single-cell analysis using a custom 96-gene panel to probe heterogeneity within the clonal CSC spheres. This microfluidic platform provides reliable and high-throughput sphere formation for CSC identification and downstream clonal analysis.
Collapse
MESH Headings
- Animals
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Cell Line, Tumor
- Female
- Humans
- Hydrogels/chemistry
- Lab-On-A-Chip Devices
- MCF-7 Cells
- Mice
- Mice, Inbred NOD
- Mice, SCID
- Microfluidics/instrumentation
- Microfluidics/methods
- Microscopy, Electron, Scanning
- Neoplastic Stem Cells/cytology
- Neoplastic Stem Cells/metabolism
- Polyhydroxyethyl Methacrylate/chemistry
- Receptors, Notch/genetics
- Receptors, Notch/metabolism
- Single-Cell Analysis/methods
- Spheroids, Cellular/cytology
- Spheroids, Cellular/metabolism
- Transplantation, Heterologous
Collapse
Affiliation(s)
- Yu-Chih Chen
- Department of Electrical Engineering and Computer Science, University of Michigan, 1301 Beal Avenue, Ann Arbor, MI 48109-2122, USA
- University of Michigan Comprehensive Cancer Center, 1500 East Medical Center Drive, Ann Arbor, MI 48109-5940, USA
| | - Patrick N. Ingram
- Department of Biomedical Engineering, University of Michigan, 2200 Bonisteel Blvd, Ann Arbor, MI 48109-2099, USA
| | - Shamileh Fouladdel
- University of Michigan Comprehensive Cancer Center, 1500 East Medical Center Drive, Ann Arbor, MI 48109-5940, USA
| | - Sean P. McDermott
- University of Michigan Comprehensive Cancer Center, 1500 East Medical Center Drive, Ann Arbor, MI 48109-5940, USA
| | - Ebrahim Azizi
- University of Michigan Comprehensive Cancer Center, 1500 East Medical Center Drive, Ann Arbor, MI 48109-5940, USA
| | - Max S. Wicha
- University of Michigan Comprehensive Cancer Center, 1500 East Medical Center Drive, Ann Arbor, MI 48109-5940, USA
| | - Euisik Yoon
- Department of Electrical Engineering and Computer Science, University of Michigan, 1301 Beal Avenue, Ann Arbor, MI 48109-2122, USA
- Department of Biomedical Engineering, University of Michigan, 2200 Bonisteel Blvd, Ann Arbor, MI 48109-2099, USA
| |
Collapse
|
361
|
Cheung PFY, Yip CW, Ng LWC, Lo KW, Chow C, Chan KF, Cheung TT, Cheung ST. Comprehensive characterization of the patient-derived xenograft and the paralleled primary hepatocellular carcinoma cell line. Cancer Cell Int 2016; 16:41. [PMID: 27279800 PMCID: PMC4898407 DOI: 10.1186/s12935-016-0322-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 06/03/2016] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is an aggressive cancer with high mortality and morbidity worldwide. The limited clinically relevant model has impeded the development of effective HCC treatment strategy. Patient-derived xenograft (PDX) models retain most of the characteristics of original tumors and were shown to be highly predictive for clinical outcomes. Notably, primary cell line models allow in-depth molecular characterization and high-throughput analysis. Combined usage of the two models would provide an excellent tool for systematic study of therapeutic strategies. Here, we comprehensively characterized the novel PDX and the paralleled primary HCC cell line model. METHODS Tumor tissues were collected from HCC surgical specimens. HCC cells were sorted for in vivo PDX and in vitro cell line establishment by the expression of hepatic cancer stem cell marker to enhance cell viability and the rate of success on subsequent culture. The PDX and its matching primary cell line were authenticated and characterized in vitro and in vivo. RESULTS Among the successful cases for generating PDXs and primary cells, HCC40 is capable for both PDX and primary cell line establishment, which were then further characterized. The novel HCC40-PDX and HCC40-CL exhibited consistent phenotypic characteristics as the original tumor in terms of HBV protein and AFP expressions. In common with HCC40-PDX, HCC40-CL was tumorigenic in immunocompromised mice. The migration ability in vitro and metastatic properties in vivo echoed the clinical feature of venous infiltration. Genetic profiling by short tandem repeat analysis and p53 mutation pattern consolidated that both the HCC40-PDX and HCC40-CL models were derived from the HCC40 clinical specimen. CONCLUSIONS The paralleled establishment of PDX and primary cell line would serve as useful models in comprehensive studies for HCC pathogenesis and therapeutics development for personalized treatment.
Collapse
Affiliation(s)
- Phyllis F Y Cheung
- Department of Surgery, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, China ; Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Hong Kong, China
| | - Chi Wai Yip
- Department of Surgery, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, China ; Department of Surgery, The University of Hong Kong, Hong Kong, China ; Division of Genomic Technologies, RIKEN Center for Life Science Technologies, Yokohama, Japan
| | - Linda W C Ng
- Department of Surgery, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, China
| | - Kwok Wai Lo
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Hong Kong, China
| | - Chit Chow
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Hong Kong, China
| | - Kui Fat Chan
- Department of Pathology, Tuen Mun Hospital, Hong Kong, China
| | - Tan To Cheung
- Department of Surgery, The University of Hong Kong, Hong Kong, China
| | - Siu Tim Cheung
- Department of Surgery, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, China ; Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
362
|
Animal Models of Uveal Melanoma: Methods, Applicability, and Limitations. BIOMED RESEARCH INTERNATIONAL 2016; 2016:4521807. [PMID: 27366747 PMCID: PMC4913058 DOI: 10.1155/2016/4521807] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 05/08/2016] [Indexed: 02/06/2023]
Abstract
Animal models serve as powerful tools for investigating the pathobiology of cancer, identifying relevant pathways, and developing novel therapeutic agents. They have facilitated rapid scientific progress in many tumor entities. However, for establishing a powerful animal model of uveal melanoma fundamental challenges remain. To date, no animal model offers specific genetic attributes as well as histologic, immunologic, and metastatic features of uveal melanoma. Syngeneic models with intraocular injection of cutaneous melanoma cells may suit best for investigating immunologic/tumor biology aspects. However, differences between cutaneous and uveal melanoma regarding genetics and metastasis remain problematic. Human xenograft models are widely used for evaluating novel therapeutics but require immunosuppression to allow tumor growth. New approaches aim to establish transgenic mouse models of spontaneous uveal melanoma which recently provided preliminary promising results. Each model provides certain benefits and may render them suitable for answering a respective scientific question. However, all existing models also exhibit relevant limitations which may have led to delayed research progress. Despite refined therapeutic options for the primary ocular tumor, patients' prognosis has not improved since the 1970s. Basic research needs to further focus on a refinement of a potent animal model which mimics uveal melanoma specific mechanisms of progression and metastasis. This review will summarise and interpret existing animal models of uveal melanoma including recent advances in the field.
Collapse
|
363
|
Abstract
Experimental oncology research and preclinical drug development both substantially require specific, clinically relevant in vitro and in vivo tumor models. The increasing knowledge about the heterogeneity of cancer requested a substantial restructuring of the test systems for the different stages of development. To be able to cope with the complexity of the disease, larger panels of patient-derived tumor models have to be implemented and extensively characterized. Together with individual genetically engineered tumor models and supported by core functions for expression profiling and data analysis, an integrated discovery process has been generated for predictive and personalized drug development.Improved “humanized” mouse models should help to overcome current limitations given by xenogeneic barrier between humans and mice. Establishment of a functional human immune system and a corresponding human microenvironment in laboratory animals will strongly support further research.Drug discovery, systems biology, and translational research are moving closer together to address all the new hallmarks of cancer, increase the success rate of drug development, and increase the predictive value of preclinical models.
Collapse
|
364
|
Neuroblastoma patient-derived orthotopic xenografts reflect the microenvironmental hallmarks of aggressive patient tumours. Cancer Lett 2016; 375:384-389. [DOI: 10.1016/j.canlet.2016.02.046] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 02/25/2016] [Accepted: 02/25/2016] [Indexed: 12/25/2022]
|
365
|
Mouse Models in Prostate Cancer Translational Research: From Xenograft to PDX. BIOMED RESEARCH INTERNATIONAL 2016; 2016:9750795. [PMID: 27294148 PMCID: PMC4887629 DOI: 10.1155/2016/9750795] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 04/21/2016] [Indexed: 12/20/2022]
Abstract
Despite the advancement of clinical and preclinical research on PCa, which resulted in the last five years in a decrement of disease incidence by 3-4%, it remains the most frequent cancer in men and the second for mortality rate. Based on this evidence we present a brief dissertation on numerous preclinical models, comparing their advantages and disadvantages; among this we report the PDX mouse models that show greater fidelity to the disease, in terms of histopathologic features of implanted tumor, gene and miRNA expression, and metastatic pattern, well describing all tumor progression stages; this characteristic encourages the translation of preclinical results. These models become particularly useful in meeting the need of new treatments identification that eradicate PCa bone metastases growing, clarifying pathway of angiogenesis, identifying castration-resistant stem-like cells, and studying the antiandrogen therapies. Also of considerable interest are the studies of 3D cell cultures derived from PDX, which have the ability to maintain PDX cell viability with continued native androgen receptor expression, also showing a differential sensitivity to drugs. 3D PDX PCa may represent a diagnostic platform for the rapid assessment of drugs and push personalized medicine. Today the development of preclinical models in vitro and in vivo is necessary in order to obtain increasingly reliable answers before reaching phase III of the drug discovery.
Collapse
|
366
|
Zhou H, Qian W, Uckun FM, Zhou Z, Wang L, Wang A, Mao H, Yang L. IGF-1 receptor targeted nanoparticles for image-guided therapy of stroma-rich and drug resistant human cancer. PROCEEDINGS OF SPIE--THE INTERNATIONAL SOCIETY FOR OPTICAL ENGINEERING 2016; 9836. [PMID: 27313332 DOI: 10.1117/12.2224914] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Low drug delivery efficiency and drug resistance from highly heterogeneous cancer cells and tumor microenvironment represent major challenges in clinical oncology. Growth factor receptor, IGF-1R, is overexpressed in both human tumor cells and tumor associated stromal cells. The level of IGF-1R expression is further up-regulated in drug resistant tumor cells. We have developed IGF-1R targeted magnetic iron oxide nanoparticles (IONPs) carrying multiple anticancer drugs into human tumors. This IGF-1R targeted theranostic nanoparticle delivery system has an iron core for non-invasive MR imaging, amphiphilic polymer coating to ensure the biocompatibility as well as for drug loading and conjugation of recombinant human IGF-1 as targeting molecules. Chemotherapy drugs, Doxorubicin (Dox), was encapsulated into the polymer coating and/or conjugated to the IONP surface by coupling with the carboxyl groups. The ability of IGF1R targeted theranostic nanoparticles to penetrate tumor stromal barrier and enhance tumor cell killing has been demonstrated in human pancreatic cancer patient tissue derived xenograft (PDX) models. Repeated systemic administrations of those IGF-1R targeted theranostic IONP carrying Dox led to breaking the tumor stromal barrier and improved therapeutic effect. Near infrared (NIR) optical and MR imaging enabled noninvasive monitoring of nanoparticle-drug delivery and therapeutic responses. Our results demonstrated that IGF-1R targeted nanoparticles carrying multiple drugs are promising combination therapy approaches for image-guided therapy of stroma-rich and drug resistant human cancer, such as pancreatic cancer.
Collapse
Affiliation(s)
- Hongyu Zhou
- Department of Surgery and Radiology, Emory University School of Medicine, Atlanta, Georgia
| | - Weiping Qian
- Department of Surgery and Radiology, Emory University School of Medicine, Atlanta, Georgia
| | - Fatih M Uckun
- University of Southern California Norris Comprehensive Cancer Center, Children's Hospital Los Angeles, Los Angeles, CA
| | - Zhiyang Zhou
- Department of Surgery and Radiology, Emory University School of Medicine, Atlanta, Georgia; Xiangya School of Medicine, Central South University, Changsha, Hunan 410008, China
| | - Liya Wang
- Department of Imaging Sciences, Emory University School of Medicine, Atlanta, Georgia
| | | | - Hui Mao
- Department of Imaging Sciences, Emory University School of Medicine, Atlanta, Georgia
| | - Lily Yang
- Department of Surgery and Radiology, Emory University School of Medicine, Atlanta, Georgia; Department of Imaging Sciences, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
367
|
Development and Preclinical Application of an Immunocompetent Transplant Model of Basal Breast Cancer with Lung, Liver and Brain Metastases. PLoS One 2016; 11:e0155262. [PMID: 27171183 PMCID: PMC4865188 DOI: 10.1371/journal.pone.0155262] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 04/26/2016] [Indexed: 01/07/2023] Open
Abstract
Triple negative breast cancer (TNBC) is an aggressive subtype of breast cancer that is associated with a poor prognosis and for which no targeted therapies currently exist. In order to improve preclinical testing for TNBC that relies primarily on using human xenografts in immunodeficient mice, we have developed a novel immunocompetent syngeneic murine tumor transplant model for basal-like triple-negative breast cancer. The C3(1)/SV40-T/t-antigen (C3(1)/Tag) mouse mammary tumor model in the FVB/N background shares important similarities with human basal-like TNBC. However, these tumors or derived cell lines are rejected when transplanted into wt FVB/N mice, likely due to the expression of SV40 T-antigen. We have developed a sub-line of mice (designated REAR mice) that carry only one copy of the C3(1)/Tag-antigen transgene resulting from a spontaneous transgene rearrangement in the original founder line. Unlike the original C3(1)/Tag mice, REAR mice do not develop mammary tumors or other phenotypes observed in the original C3(1)/Tag transgenic mice. REAR mice are more immunologically tolerant to SV40 T-antigen driven tumors and cell lines in an FVB/N background (including prostate tumors from TRAMP mice), but are otherwise immunologically intact. This transplant model system offers the ability to synchronously implant the C3(1)/Tag tumor-derived M6 cell line or individual C3(1)/Tag tumors from various stages of tumor development into the mammary fat pads or tail veins of REAR mice. C3(1)/Tag tumors or M6 cells implanted into the mammary fat pads spontaneously metastasize at a high frequency to the lung and liver. M6 cells injected by tail vein can form brain metastases. We demonstrate that irradiated M6 tumor cells or the same cells expressing GM-CSF can act as a vaccine to retard tumor growth of implanted tumor cells in the REAR model. Preclinical studies performed in animals with an intact immune system should more authentically replicate treatment responses in human patients.
Collapse
|
368
|
Guastella AR, Michelhaugh SK, Klinger NV, Kupsky WJ, Polin LA, Muzik O, Juhász C, Mittal S. Tryptophan PET Imaging of the Kynurenine Pathway in Patient-Derived Xenograft Models of Glioblastoma. Mol Imaging 2016; 15:15/0/1536012116644881. [PMID: 27151136 PMCID: PMC4887573 DOI: 10.1177/1536012116644881] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 03/18/2016] [Indexed: 11/17/2022] Open
Abstract
Increasing evidence demonstrates the immunosuppressive kynurenine pathway's (KP) role in the pathophysiology of human gliomas. To study the KP in vivo, we used the noninvasive molecular imaging tracer α-[(11)C]-methyl-l-tryptophan (AMT). The AMT-positron emission tomography (PET) has shown high uptake in high-grade gliomas and predicted survival in patients with recurrent glioblastoma (GBM). We generated patient-derived xenograft (PDX) models from dissociated cells, or tumor fragments, from 5 patients with GBM. Mice bearing subcutaneous tumors were imaged with AMT-PET, and tumors were analyzed to detect the KP enzymes indoleamine 2,3-dioxygenase (IDO) 1, IDO2, tryptophan 2,3-dioxygenase, kynureninase, and kynurenine 3-monooxygenase. Overall, PET imaging showed robust tumoral AMT uptake in PDX mice with prolonged tracer accumulation over 60 minutes, consistent with AMT trapping seen in humans. Immunostained tumor tissues demonstrated positive detection of multiple KP enzymes. Furthermore, intracranial implantation of GBM cells was performed with imaging at both 9 and 14 days postimplant, with a marked increase in AMT uptake at 14 days and a corresponding high level of tissue immunostaining for KP enzymes. These results indicate that our PDX mouse models recapitulate human GBM, including aberrant tryptophan metabolism, and offer an in vivo system for development of targeted therapeutics for patients with GBM.
Collapse
Affiliation(s)
- Anthony R Guastella
- Department of Neurosurgery, Wayne State University, Detroit, MI, USA Department of Oncology, Wayne State University, Detroit, MI, USA
| | | | - Neil V Klinger
- Department of Neurosurgery, Wayne State University, Detroit, MI, USA
| | - William J Kupsky
- Department of Pathology, Wayne State University, Detroit, MI, USA Karmanos Cancer Institute, Detroit, MI, USA
| | - Lisa A Polin
- Department of Pathology, Wayne State University, Detroit, MI, USA Karmanos Cancer Institute, Detroit, MI, USA
| | - Otto Muzik
- Department of Pediatrics, Wayne State University, Detroit, MI, USA Department of Radiology, Wayne State University, Detroit, MI, USA PET Center and Translational Imaging Laboratory, Children's Hospital of Michigan, Detroit, MI, USA
| | - Csaba Juhász
- Karmanos Cancer Institute, Detroit, MI, USA Department of Pediatrics, Wayne State University, Detroit, MI, USA PET Center and Translational Imaging Laboratory, Children's Hospital of Michigan, Detroit, MI, USA Department of Neurology, Wayne State University, Detroit, MI, USA
| | - Sandeep Mittal
- Department of Neurosurgery, Wayne State University, Detroit, MI, USA Department of Oncology, Wayne State University, Detroit, MI, USA Karmanos Cancer Institute, Detroit, MI, USA
| |
Collapse
|
369
|
Correlation between tumor engraftment in patient-derived xenograft models and clinical outcomes in colorectal cancer patients. Oncotarget 2016; 6:16059-68. [PMID: 25965827 PMCID: PMC4599256 DOI: 10.18632/oncotarget.3863] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Accepted: 04/02/2015] [Indexed: 02/07/2023] Open
Abstract
Despite numerous studies involving patient-derived xenograft (PDX) models, few studies have investigated the relationship between the ability of the tumor to engraft (tumorigenicity) and the clinical features of colorectal cancer (CRC). The aim of this study was to determine whether tumorigenicity correlates with clinical outcomes of CRC patients. We included 241 CRC patients who underwent radical surgery from 2010 to 2013. PDX models were established by implanting tumor fragments obtained from these patients into the subcutaneous layer of immunodeficient mice. Xenografts were successfully established from 62.2%. Successful engraftment was associated with advanced stage (p < 0.001) and moderate/poor differentiation (p = 0.029). Three-year disease-free survival (DFS) rates were lower for patients with tumorigenicity (p = 0.011). In stage III patients, tumorigenicity was an independent predictor of poor DFS (p = 0.034). In addition, mutation of TP53 was most frequently detected in stage III patients with tumorigenicity. Two models of stage IV disease without KRAS mutations showed high sensitivity to EGFR-targeted agents, while none of the models with KRAS mutations showed high sensitivity. In conclusion, PDX models may provide an effective preclinical tool for predicting cancer progression and could be used to further genomic and pharmacologic research on personalized treatments.
Collapse
|
370
|
Shi C, Wu JB, Pan D. Review on near-infrared heptamethine cyanine dyes as theranostic agents for tumor imaging, targeting, and photodynamic therapy. JOURNAL OF BIOMEDICAL OPTICS 2016; 21:50901. [PMID: 27165449 DOI: 10.1117/1.jbo.21.5.050901] [Citation(s) in RCA: 178] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 04/20/2016] [Indexed: 05/21/2023]
Abstract
A class of near-infrared fluorescence (NIRF) heptamethine cyanine dyes that are taken up and accumulated specifically in cancer cells without chemical conjugation have recently emerged as promising tools for tumor imaging and targeting. In addition to their fluorescence and nuclear imaging-based tumor-imaging properties, these dyes can be developed as drug carriers to safely deliver chemotherapy drugs to tumors. They can also be used as effective agents for photodynamic therapy with remarkable tumoricidal activity via photodependent cytotoxic activity. The preferential uptake of dyes into cancer but not normal cells is co-operatively mediated by the prevailing activation of a group of organic anion-transporting polypeptides on cancer cell membranes, as well as tumor hypoxia and increased mitochondrial membrane potential in cancer cells. Such mechanistic explorations have greatly advanced the current application and future development of NIRF dyes and their derivatives as anticancer theranostic agents. This review summarizes current knowledge and emerging advances in NIRF dyes, including molecular characterization, photophysical properties, multimodal development and uptake mechanisms, and their growing potential for preclinical and clinical use.
Collapse
Affiliation(s)
- Changhong Shi
- Fourth Military Medical University, Laboratory Animal Center, 169 West Changle Road, Xi'an, Shaanxi 710032, China
| | - Jason Boyang Wu
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Uro-Oncology Research Program, Department of Medicine, Los Angeles, California 90048, United States
| | - Dongfeng Pan
- University of Virginia, Department of Radiology, Charlottesville, Virginia 22908, United States
| |
Collapse
|
371
|
Kim KT, Lee HW, Lee HO, Song HJ, Jeong DE, Shin S, Kim H, Shin Y, Nam DH, Jeong BC, Kirsch DG, Joo KM, Park WY. Application of single-cell RNA sequencing in optimizing a combinatorial therapeutic strategy in metastatic renal cell carcinoma. Genome Biol 2016; 17:80. [PMID: 27139883 PMCID: PMC4852434 DOI: 10.1186/s13059-016-0945-9] [Citation(s) in RCA: 149] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 04/11/2016] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Intratumoral heterogeneity hampers the success of marker-based anticancer treatment because the targeted therapy may eliminate a specific subpopulation of tumor cells while leaving others unharmed. Accordingly, a rational strategy minimizing survival of the drug-resistant subpopulation is essential to achieve long-term therapeutic efficacy. RESULTS Using single-cell RNA sequencing (RNA-seq), we examine the intratumoral heterogeneity of a pair of primary renal cell carcinoma and its lung metastasis. Activation of drug target pathways demonstrates considerable variability between the primary and metastatic sites, as well as among individual cancer cells within each site. Based on the prediction of multiple drug target pathway activation, we derive a combinatorial regimen co-targeting two mutually exclusive pathways for the metastatic cancer cells. This combinatorial strategy shows significant increase in the treatment efficacy over monotherapy in the experimental validation using patient-derived xenograft platforms in vitro and in vivo. CONCLUSIONS Our findings demonstrate the investigational application of single-cell RNA-seq in the design of an anticancer regimen. The approach may overcome intratumoral heterogeneity which hampers the success of precision medicine.
Collapse
Affiliation(s)
- Kyu-Tae Kim
- Samsung Genome Institute, Samsung Medical Center, Seoul, South Korea
| | - Hye Won Lee
- Institute for Future Medicine, Samsung Medical Center, Seoul, South Korea
| | - Hae-Ock Lee
- Samsung Genome Institute, Samsung Medical Center, Seoul, South Korea.,Departments of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Hye Jin Song
- Departments of Anatomy and Cell Biology, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Da Eun Jeong
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University, Seoul, South Korea
| | - Sang Shin
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University, Seoul, South Korea
| | - Hyunho Kim
- School of Mechanical Engineering, Korea University, Seoul, South Korea
| | - Yoojin Shin
- School of Mechanical Engineering, Korea University, Seoul, South Korea
| | - Do-Hyun Nam
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University, Seoul, South Korea.,Departments of Neurosurgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Byong Chang Jeong
- Departments of Urology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - David G Kirsch
- Departments of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, USA.,Departments of Radiation Oncology, Duke University Medical Center, Durham, NC, USA
| | - Kyeung Min Joo
- Departments of Anatomy and Cell Biology, Sungkyunkwan University School of Medicine, Seoul, South Korea. .,Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University, Seoul, South Korea.
| | - Woong-Yang Park
- Samsung Genome Institute, Samsung Medical Center, Seoul, South Korea. .,Departments of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Seoul, South Korea. .,Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University, Seoul, South Korea.
| |
Collapse
|
372
|
Jiang J, Wang DD, Yang M, Chen D, Pang L, Guo S, Cai J, Wery JP, Li L, Li HQ, Lin PP. Comprehensive characterization of chemotherapeutic efficacy on metastases in the established gastric neuroendocrine cancer patient derived xenograft model. Oncotarget 2016; 6:15639-51. [PMID: 25909226 PMCID: PMC4558176 DOI: 10.18632/oncotarget.3712] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2015] [Accepted: 03/10/2015] [Indexed: 01/10/2023] Open
Abstract
The HuPrime® human gastric neuroendocrine carcinoma derived xenograft model GA0087 was established in this study. GA0087 PDX model showed high gene expression of vascular endothelial growth factors (VEGF)-A and B, and high potential of lung metastasis. Circulating tumor cells (CTCs) with either large or small size, circulating tumor microemboli (CTM) and lung metastatic lesions were detected in GA0087 PDX mice. The number of CTC correlated to the number of metastatic nodules in lung. Both primary tumor growth and metastasis in terms of the number of dynamically monitored CTCs and metastatic nodules were effectively suppressed by Cisplatin. Diverse subtypes of CTCs in the context of sensitivity to Cisplatin were specifically identified by subtraction enrichment (SE) integrated with in situ Phenotyping of cytokeratin 18 (CK18) and Karyotyping of chromosome 8 (in situ PK CTC by CK-iFISH). All the CK18-/diploid and majority of CK18+/diploid CTC subtypes were chemosensitive, whereas a higher percentage of CK18+/multiploid subtype of CTC were Cisplatin-insensitive. Combined histopathological examination of metastatic lesion and in situ PK CTC in a metastatic PDX (mPDX) tumor model are of particular significance, and may provide an unique and robust platform for cancer research as well as pre-clinical evaluation of therapeutic efficacy of new anti-cancer drugs.
Collapse
Affiliation(s)
| | | | | | - Dawei Chen
- Crown Bioscience, Santa Clara, California, USA
| | - Liang Pang
- Crown Bioscience, Santa Clara, California, USA
| | - Sheng Guo
- Crown Bioscience, Santa Clara, California, USA
| | - Jie Cai
- Crown Bioscience, Santa Clara, California, USA
| | | | - Linda Li
- Cytelligen, San Diego, California, USA
| | - Henry Qixiang Li
- Crown Bioscience, Santa Clara, California, USA.,State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, China
| | | |
Collapse
|
373
|
Usary J, Darr DB, Pfefferle A, Perou CM. Overview of Genetically Engineered Mouse Models of Distinct Breast Cancer Subtypes. CURRENT PROTOCOLS IN PHARMACOLOGY 2016; 72:14.38.1-14.38.11. [PMID: 26995547 PMCID: PMC4826719 DOI: 10.1002/0471141755.ph1438s72] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Advances in the screening of new therapeutic options have significantly reduced the breast cancer death rate over the last decade. Despite these advances, breast cancer remains the second leading cause of cancer death among women. This is due in part to the complexity of the disease, which is characterized by multiple subtypes that are driven by different genetic mechanisms and that likely arise from different cell types of origin. Because these differences often drive treatment options and outcomes, it is important to select relevant preclinical model systems to study new therapeutic interventions and tumor biology. Described in this unit are the characteristics and applications of validated genetically engineered mouse models (GEMMs) of basal-like, luminal, and claudin-low human subtypes of breast cancer. These different subtypes have different clinical outcomes and require different treatment strategies. These GEMMs can be considered faithful surrogates of their human disease counterparts. They represent alternative preclinical tumor models to cell line and patient-derived xenografts for preclinical drug discovery and tumor biology studies.
Collapse
Affiliation(s)
- Jerry Usary
- Dept. of Genetics, The University of North Carolina at Chapel Hill, 450 West Drive, Chapel Hill NC 27599, Tel: 919-843-5740
| | - David Brian Darr
- Dept. of Genetics, The University of North Carolina at Chapel Hill, 450 West Drive, Chapel Hill NC 27599, Tel: 919-843-5740
| | - Adam Pfefferle
- Dept. of Genetics, The University of North Carolina at Chapel Hill, 450 West Drive, Chapel Hill NC 27599, Tel: 919-843-5740
| | - Charles M. Perou
- Dept. of Genetics, The University of North Carolina at Chapel Hill, 450 West Drive, Chapel Hill NC 27599, Tel: 919-843-5740
| |
Collapse
|
374
|
Izumchenko E, Meir J, Bedi A, Wysocki PT, Hoque MO, Sidransky D. Patient-derived xenografts as tools in pharmaceutical development. Clin Pharmacol Ther 2016; 99:612-21. [PMID: 26874468 DOI: 10.1002/cpt.354] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 02/20/2016] [Accepted: 02/11/2016] [Indexed: 12/16/2022]
Abstract
Successful drug development in oncology is grossly suboptimal, manifested by the very low percentage of new agents being developed that ultimately succeed in clinical approval. This poor success is in part due to the inability of standard cell-line xenograft models to accurately predict clinical success and to tailor chemotherapy specifically to a group of patients more likely to benefit from the therapy. Patient-derived xenografts (PDXs) maintain the histopathological architecture and molecular features of human tumors, and offer a potential solution to maximize drug development success and ultimately generate better outcomes for patients. Although imperfect in mimicking all aspects of human cancer, PDXs are a more predictable platform for preclinical evaluation of treatment effect and in selected cases can guide therapeutic decision making in the clinic. This article summarizes the current status of PDX models, challenges associated with modeling human cancer, and various approaches that have been applied to overcome these challenges and improve the clinical relevance of PDX cancer models.
Collapse
Affiliation(s)
- E Izumchenko
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - J Meir
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - A Bedi
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - P T Wysocki
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - M O Hoque
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - D Sidransky
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
375
|
Huang Y, Cheng C, Zhang C, Zhang Y, Chen M, Strand DW, Jiang M. Advances in prostate cancer research models: From transgenic mice to tumor xenografting models. Asian J Urol 2016; 3:64-74. [PMID: 29264167 PMCID: PMC5730804 DOI: 10.1016/j.ajur.2016.02.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Revised: 01/01/2016] [Accepted: 02/02/2016] [Indexed: 12/17/2022] Open
Abstract
The identification of the origin and molecular characteristics of prostate cancer (PCa) has crucial implications for personalized treatment. The development of effective treatments for PCa has been limited; however, the recent establishment of several transgenic mouse lines and/or xenografting models is better reflecting the disease in vivo. With appropriate models, valuable tools for elucidating the functions of specific genes have gone deep into prostate development and carcinogenesis. In the present review, we summarize a number of important PCa research models established in our laboratories (PSA-Cre-ERT2/PTEN transgenic mouse models, AP-OX model, tissue recombination-xenografting models and PDX models), which represent advances of translational models from transgenic mouse lines to human tumor xenografting. Better understanding of the developments of these models will offer new insights into tumor progression and may help explain the functional significance of genetic variations in PCa. Additionally, this understanding could lead to new modes for curing PCa based on their particular biological phenotypes.
Collapse
Affiliation(s)
- Yuejiao Huang
- Department of Oncology, Affiliated Cancer Hospital of Nantong University, Nantong, Jiangsu, China
| | - Chun Cheng
- Department of Immunology, Nantong University School of Medicine, Nantong, Jiangsu, China
| | - Chong Zhang
- Laboratory of Nuclear Receptors and Cancer Research, Center for Basic Medical Research, Nantong University School of Medicine, Nantong, Jiangsu, China
| | - Yonghui Zhang
- Laboratory of Nuclear Receptors and Cancer Research, Center for Basic Medical Research, Nantong University School of Medicine, Nantong, Jiangsu, China
| | - Miaomiao Chen
- Laboratory of Nuclear Receptors and Cancer Research, Center for Basic Medical Research, Nantong University School of Medicine, Nantong, Jiangsu, China
| | - Douglas W Strand
- Department of Urology, UT Southernwestern Medical Center, Dallas, TX, USA
| | - Ming Jiang
- Laboratory of Nuclear Receptors and Cancer Research, Center for Basic Medical Research, Nantong University School of Medicine, Nantong, Jiangsu, China.,Institute of Medicine and Public Health, Division of Epidemiology, Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
376
|
Ludwig ML, Birkeland AC, Hoesli R, Swiecicki P, Spector ME, Brenner JC. Changing the paradigm: the potential for targeted therapy in laryngeal squamous cell carcinoma. Cancer Biol Med 2016; 13:87-100. [PMID: 27144065 PMCID: PMC4850131 DOI: 10.28092/j.issn.2095-3941.2016.0010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 02/17/2016] [Indexed: 01/05/2023] Open
Abstract
Laryngeal squamous cell carcinoma (LSCC) remains a highly morbid and fatal disease. Historically, it has been a model example for organ preservation and treatment stratification paradigms. Unfortunately, survival for LSCC has stagnated over the past few decades. As the era of next-generation sequencing and personalized treatment for cancer approaches, LSCC may be an ideal disease for consideration of further treatment stratification and personalization. Here, we will discuss the important history of LSCC as a model system for organ preservation, unique and potentially targetable genetic signatures of LSCC, and methods for bringing stratified, personalized treatment strategies to the 21(st) century.
Collapse
Affiliation(s)
- Megan L. Ludwig
- Department of Otolaryngology, Head and Neck Surgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, MI 48109, USA
| | - Andrew C. Birkeland
- Department of Otolaryngology, Head and Neck Surgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Rebecca Hoesli
- Department of Otolaryngology, Head and Neck Surgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Paul Swiecicki
- Department of Hematology Oncology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Matthew E. Spector
- Department of Otolaryngology, Head and Neck Surgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - J. Chad Brenner
- Department of Otolaryngology, Head and Neck Surgery, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, MI 48109, USA
- Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| |
Collapse
|
377
|
Establishment and characterisation of patient-derived xenografts as paraclinical models for gastric cancer. Sci Rep 2016; 6:22172. [PMID: 26926953 PMCID: PMC4772087 DOI: 10.1038/srep22172] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 02/09/2016] [Indexed: 12/14/2022] Open
Abstract
The patient-derived xenograft (PDX) model is emerging as a promising translational platform to duplicate the characteristics of tumours. However, few studies have reported detailed histological and genomic analyses for model fidelity and for factors affecting successful model establishment of gastric cancer. Here, we generated PDX tumours surgically-derived from 62 gastric cancer patients. Fifteen PDX models were successfully established (24.2%, 15/62) and passaged to maintain tumours in immune-compromised mice. Diffuse type and low tumour cell percentage were negatively correlated with success rates (p = 0.005 and p = 0.025, respectively), while reducing ex vivo and overall procedure times were positively correlated with success rates (p = 0.003 and p = 0.01, respectively). The histology and genetic characteristics of PDX tumour models were stable over subsequent passages. Lymphoma transformation occurred in five cases (33.3%, 5/15), and all were in the NOG mouse, with none in the nude mouse. Together, the present study identified Lauren classification, tumour cell percentages, and ex vivo times along with overall procedure times, as key determinants for successful PDX engraftment. Furthermore, genetic and histological characteristics were highly consistent between primary and PDX tumours, which provide realistic paraclinical models, enabling personalised development of treatment options for gastric cancer.
Collapse
|
378
|
Roscilli G, De Vitis C, Ferrara FF, Noto A, Cherubini E, Ricci A, Mariotta S, Giarnieri E, Giovagnoli MR, Torrisi MR, Bergantino F, Costantini S, Fenizia F, Lambiase M, Aurisicchio L, Normanno N, Ciliberto G, Mancini R. Human lung adenocarcinoma cell cultures derived from malignant pleural effusions as model system to predict patients chemosensitivity. J Transl Med 2016; 14:61. [PMID: 26928703 PMCID: PMC4772534 DOI: 10.1186/s12967-016-0816-x] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 02/13/2016] [Indexed: 12/31/2022] Open
Abstract
Background Lung cancer is the leading cause of cancer related deaths and Malignant Pleural Effusion (MPE) is a frequent complication. Current therapies suffer from lack of efficacy in a great percentage of cases, especially when cancer is diagnosed at a late stage. Moreover patients’ responses vary and the outcome is unpredictable. Therefore, the identification of patients who will benefit most of chemotherapy treatment is important for accurate prognostication and better outcome. In this study, using malignant pleural effusions (MPE) from non-small cell lung cancer (NSCLC) patients, we established a collection of patient-derived Adenocarcinoma cultures which were characterized for their sensitivity to chemotherapeutic drugs used in the clinical practice. Methods Tumor cells present in MPEs of patients with NSCLC were isolated by density gradient centrifugation, placed in culture and genotyped by next generation sequencing. In a subset of cases patient derived xenografts (PDX) were obtained upon tumor cell inoculation in rag2/IL2 knock-out mice. Isolated primary cultures were characterized and tested for drug sensitivity by in vitro proliferation assays. Additivity, antagonism or synergy for combinatorial treatments were determined by analysis with the Calcusyn software. Results We have optimized isolation procedures and culture conditions to expand in vitro primary cultures from Malignant Pleural Effusions (MPEs) of patients affected by lung adenocarcinomas, the most frequent form of non small cell lung cancer. Using this approach we have been able to establish 16 primary cultures from MPEs. Cells were banked at low passages and were characterized for their mutational pattern by next generation sequencing for most common driver mutations in lung cancer. Moreover, amplified cultures were shown to engraft with high efficiency when injected in immunocompromised mice. Cancer cell sensitivity to drugs used in standard chemotherapy regimens was assessed either individually or in combination. Differential chemosensitivity and different mutation profiles were observed which suggests that this isolation method could provide a platform for predicting the efficacy of chemotherapy in the clinical setting. Most importantly for six patients it was possible to establish a correlation between drug response in vitro and response to therapy in the clinic. Conclusions Results obtained using primary cultured cells from MPEs underscore the heterogeneity of NSCLC in advanced stage as indicated by drug response and mutation profile. Comparison of data obtained from in vitro assays with patients’ responses to therapy leads to the conclusion that this strategy may provide a potentially useful approach for evaluating individual chemosensitivity profile and tailor the therapy accordingly. Furthermore, combining MPE-derived primary cultures with their genomic testing allows to identify patients eligible to trials with novel targeted agents. Electronic supplementary material The online version of this article (doi:10.1186/s12967-016-0816-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Giuseppe Roscilli
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Rome, Italy. .,Takis srl, Rome, Italy.
| | - Claudia De Vitis
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Rome, Italy. .,Laboratory of Research and Diagnostics, Department of Surgery "P.Valdoni", Sapienza University of Rome, Rome, Italy.
| | | | - Alessia Noto
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Rome, Italy. .,Laboratory of Research and Diagnostics, Department of Surgery "P.Valdoni", Sapienza University of Rome, Rome, Italy.
| | - Emanuela Cherubini
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Rome, Italy.
| | - Alberto Ricci
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Rome, Italy. .,Azienda Ospedaliera S. Andrea, Rome, Italy.
| | - Salvatore Mariotta
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Rome, Italy. .,Azienda Ospedaliera S. Andrea, Rome, Italy.
| | - Enrico Giarnieri
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Rome, Italy. .,Azienda Ospedaliera S. Andrea, Rome, Italy.
| | - Maria Rosaria Giovagnoli
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Rome, Italy. .,Azienda Ospedaliera S. Andrea, Rome, Italy.
| | - Maria Rosaria Torrisi
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Rome, Italy. .,Azienda Ospedaliera S. Andrea, Rome, Italy.
| | | | - Susan Costantini
- IRCCS Istituto Nazionale Tumori, Fondazione "G. Pascale", Naples, Italy.
| | - Francesca Fenizia
- IRCCS Istituto Nazionale Tumori, Fondazione "G. Pascale", Naples, Italy.
| | - Matilde Lambiase
- IRCCS Istituto Nazionale Tumori, Fondazione "G. Pascale", Naples, Italy.
| | | | - Nicola Normanno
- IRCCS Istituto Nazionale Tumori, Fondazione "G. Pascale", Naples, Italy.
| | - Gennaro Ciliberto
- IRCCS Istituto Nazionale Tumori, Fondazione "G. Pascale", Naples, Italy.
| | - Rita Mancini
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Rome, Italy. .,Laboratory of Research and Diagnostics, Department of Surgery "P.Valdoni", Sapienza University of Rome, Rome, Italy.
| |
Collapse
|
379
|
Hayakawa Y, Kawada M, Nishikawa H, Ochiya T, Saya H, Seimiya H, Yao R, Hayashi M, Kai C, Matsuda A, Naoe T, Ohtsu A, Okazaki T, Saji H, Sata M, Sugimura H, Sugiyama Y, Toi M, Irimura T. Report on the use of non-clinical studies in the regulatory evaluation of oncology drugs. Cancer Sci 2016; 107:189-202. [PMID: 26919617 PMCID: PMC4768389 DOI: 10.1111/cas.12857] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Revised: 12/04/2015] [Accepted: 12/04/2015] [Indexed: 01/04/2023] Open
Abstract
Non‐clinical studies are necessary at each stage of the development of oncology drugs. Many experimental cancer models have been developed to investigate carcinogenesis, cancer progression, metastasis, and other aspects in cancer biology and these models turned out to be useful in the efficacy evaluation and the safety prediction of oncology drugs. While the diversity and the degree of engagement in genetic changes in the initiation of cancer cell growth and progression are widely accepted, it has become increasingly clear that the roles of host cells, tissue microenvironment, and the immune system also play important roles in cancer. Therefore, the methods used to develop oncology drugs should continuously be revised based on the advances in our understanding of cancer. In this review, we extensively summarize the effective use of those models, their advantages and disadvantages, ranges to be evaluated and limitations of the models currently used for the development and for the evaluation of oncology drugs. This review summarizes the effective use of animal models, their advantages and disadvantages, ranges to be evaluated and limitations of the models currently used for the development and for the evaluation of oncology drugs.
Collapse
Affiliation(s)
- Yoshihiro Hayakawa
- Subcommittee on Non-clinical Studies, The Science Board to the Pharmaceuticals and Medical Devices Agency, Tokyo, Japan.,Division of Pathogenic Biochemistry, Department of Bioscience, Institute of Natural Medicine, University of Toyama, Toyama, Japan
| | - Manabu Kawada
- Subcommittee on Non-clinical Studies, The Science Board to the Pharmaceuticals and Medical Devices Agency, Tokyo, Japan.,Institute of Microbial Chemistry, Microbial Chemistry Research Foundation, Numazu-shi, Japan
| | - Hiroyoshi Nishikawa
- Subcommittee on Non-clinical Studies, The Science Board to the Pharmaceuticals and Medical Devices Agency, Tokyo, Japan.,Division of Cancer Immunology, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Chiba, Japan
| | - Takahiro Ochiya
- Subcommittee on Non-clinical Studies, The Science Board to the Pharmaceuticals and Medical Devices Agency, Tokyo, Japan.,Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, Tokyo, Japan
| | - Hideyuki Saya
- Subcommittee on Non-clinical Studies, The Science Board to the Pharmaceuticals and Medical Devices Agency, Tokyo, Japan.,Division of Gene Regulation, Institute for Advanced Medical Research, School of Medicine, Keio University, Tokyo, Japan
| | - Hiroyuki Seimiya
- Subcommittee on Non-clinical Studies, The Science Board to the Pharmaceuticals and Medical Devices Agency, Tokyo, Japan.,Division of Molecular Biotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Ryoji Yao
- Subcommittee on Non-clinical Studies, The Science Board to the Pharmaceuticals and Medical Devices Agency, Tokyo, Japan.,Division of Cell Biology, Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Masahiro Hayashi
- Subcommittee on Non-clinical Studies, The Science Board to the Pharmaceuticals and Medical Devices Agency, Tokyo, Japan.,Department of Pharmacy, Toranomon Hospital, Tokyo, Japan
| | - Chieko Kai
- Subcommittee on Non-clinical Studies, The Science Board to the Pharmaceuticals and Medical Devices Agency, Tokyo, Japan.,Laboratory Animal Research Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Akira Matsuda
- Subcommittee on Non-clinical Studies, The Science Board to the Pharmaceuticals and Medical Devices Agency, Tokyo, Japan.,Department of Medicinal Chemistry, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Tomoki Naoe
- Subcommittee on Non-clinical Studies, The Science Board to the Pharmaceuticals and Medical Devices Agency, Tokyo, Japan.,National Hospital Organization Nagoya Medical Center, Nagoya, Japan
| | - Atsushi Ohtsu
- Subcommittee on Non-clinical Studies, The Science Board to the Pharmaceuticals and Medical Devices Agency, Tokyo, Japan.,Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Chiba, Japan
| | - Taku Okazaki
- Subcommittee on Non-clinical Studies, The Science Board to the Pharmaceuticals and Medical Devices Agency, Tokyo, Japan.,Division of Immune Regulation, Institute for Genome Research, Tokushima University, Tokushima, Japan
| | - Hideo Saji
- Subcommittee on Non-clinical Studies, The Science Board to the Pharmaceuticals and Medical Devices Agency, Tokyo, Japan.,Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Masataka Sata
- Subcommittee on Non-clinical Studies, The Science Board to the Pharmaceuticals and Medical Devices Agency, Tokyo, Japan.,Department of Cardiovascular Medicine, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Haruhiko Sugimura
- Subcommittee on Non-clinical Studies, The Science Board to the Pharmaceuticals and Medical Devices Agency, Tokyo, Japan.,Department of Tumor Pathology, Hamamatsu University School of Medicine, Shizuoka, Japan
| | - Yuichi Sugiyama
- Subcommittee on Non-clinical Studies, The Science Board to the Pharmaceuticals and Medical Devices Agency, Tokyo, Japan.,Sugiyama Laboratory, RIKEN Innovation Center, RIKEN Cluster for Industry Partnerships, Kanagawa, Japan
| | - Masakazu Toi
- Subcommittee on Non-clinical Studies, The Science Board to the Pharmaceuticals and Medical Devices Agency, Tokyo, Japan.,Department of Breast Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tatsuro Irimura
- Subcommittee on Non-clinical Studies, The Science Board to the Pharmaceuticals and Medical Devices Agency, Tokyo, Japan.,Juntendo University School of Medicine, Tokyo, Japan
| |
Collapse
|
380
|
Jain S, Aresu L, Comazzi S, Shi J, Worrall E, Clayton J, Humphries W, Hemmington S, Davis P, Murray E, Limeneh AA, Ball K, Ruckova E, Muller P, Vojtesek B, Fahraeus R, Argyle D, Hupp TR. The Development of a Recombinant scFv Monoclonal Antibody Targeting Canine CD20 for Use in Comparative Medicine. PLoS One 2016; 11:e0148366. [PMID: 26894679 PMCID: PMC4760772 DOI: 10.1371/journal.pone.0148366] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Accepted: 01/19/2016] [Indexed: 01/08/2023] Open
Abstract
Monoclonal antibodies are leading agents for therapeutic treatment of human diseases, but are limited in use by the paucity of clinically relevant models for validation. Sporadic canine tumours mimic the features of some human equivalents. Developing canine immunotherapeutics can be an approach for modeling human disease responses. Rituximab is a pioneering agent used to treat human hematological malignancies. Biologic mimics that target canine CD20 are just being developed by the biotechnology industry. Towards a comparative canine-human model system, we have developed a novel anti-CD20 monoclonal antibody (NCD1.2) that binds both human and canine CD20. NCD1.2 has a sub-nanomolar Kd as defined by an octet red binding assay. Using FACS, NCD1.2 binds to clinically derived canine cells including B-cells in peripheral blood and in different histotypes of B-cell lymphoma. Immunohistochemical staining of canine tissues indicates that the NCD1.2 binds to membrane localized cells in Diffuse Large B-cell lymphoma, Marginal Zone Lymphoma, and other canine B-cell lymphomas. We cloned the heavy and light chains of NCD1.2 from hybridomas to determine whether active scaffolds can be acquired as future biologics tools. The VH and VL genes from the hybridomas were cloned using degenerate primers and packaged as single chains (scFv) into a phage-display library. Surprisingly, we identified two scFv (scFv-3 and scFv-7) isolated from the hybridoma with bioactivity towards CD20. The two scFv had identical VH genes but different VL genes and identical CDR3s, indicating that at least two light chain mRNAs are encoded by NCD1.2 hybridoma cells. Both scFv-3 and scFv-7 were cloned into mammalian vectors for secretion in CHO cells and the antibodies were bioactive towards recombinant CD20 protein or peptide. The scFv-3 and scFv-7 were cloned into an ADEPT-CPG2 bioconjugate vector where bioactivity was retained when expressed in bacterial systems. These data identify a recombinant anti-CD20 scFv that might form a useful tool for evaluation in bioconjugate-directed anti-CD20 immunotherapies in comparative medicine.
Collapse
Affiliation(s)
- Saurabh Jain
- University of Edinburgh, Institute of Genetic and Molecular Medicine and School of Veterinary Medicine, Edinburgh, EH4 2XR, United Kingdom
| | - Luca Aresu
- Dipartimento di Biomedicina Comparata e Alimentazione (BCA) Department of Comparative Biomedicine and Food Science, Università di Padova 35020 Legnaro (PD), Italy
| | - Stefano Comazzi
- Dipartimento di Scienze Veterinarie e Sanità Pubblica, Università degli Studi di Milano, via Celoria 10, 20133 Milano, Italy
| | - Jianguo Shi
- University of Edinburgh, Institute of Genetic and Molecular Medicine and School of Veterinary Medicine, Edinburgh, EH4 2XR, United Kingdom
| | - Erin Worrall
- University of Edinburgh, Institute of Genetic and Molecular Medicine and School of Veterinary Medicine, Edinburgh, EH4 2XR, United Kingdom
| | - John Clayton
- Mologic, Ltd, Bedford Technology Park, Thurleigh, Bedford, MK44 2YP, United Kingdom
| | - William Humphries
- Mologic, Ltd, Bedford Technology Park, Thurleigh, Bedford, MK44 2YP, United Kingdom
| | - Sandra Hemmington
- Mologic, Ltd, Bedford Technology Park, Thurleigh, Bedford, MK44 2YP, United Kingdom
| | - Paul Davis
- Mologic, Ltd, Bedford Technology Park, Thurleigh, Bedford, MK44 2YP, United Kingdom
| | - Euan Murray
- University of Edinburgh, Institute of Genetic and Molecular Medicine and School of Veterinary Medicine, Edinburgh, EH4 2XR, United Kingdom
- INSERM Unité 940, Institut de Génétique Moléculaire, Université Paris 7, Hôpital St Louis, 27 rue Juliette Dodu, Paris, France
| | - Asmare A. Limeneh
- Bahit Dar University College of Medicine and Health Sciences Department of Medical Biochemistry and Molecular Biology, Bahir Dar, Ethiopia
| | - Kathryn Ball
- University of Edinburgh, Institute of Genetic and Molecular Medicine and School of Veterinary Medicine, Edinburgh, EH4 2XR, United Kingdom
| | - Eva Ruckova
- Regional Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, 656 53 Brno, Czech Republic
| | - Petr Muller
- Regional Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, 656 53 Brno, Czech Republic
| | - Borek Vojtesek
- Regional Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, 656 53 Brno, Czech Republic
| | - Robin Fahraeus
- INSERM Unité 940, Institut de Génétique Moléculaire, Université Paris 7, Hôpital St Louis, 27 rue Juliette Dodu, Paris, France
| | - David Argyle
- University of Edinburgh, Institute of Genetic and Molecular Medicine and School of Veterinary Medicine, Edinburgh, EH4 2XR, United Kingdom
| | - Ted R. Hupp
- University of Edinburgh, Institute of Genetic and Molecular Medicine and School of Veterinary Medicine, Edinburgh, EH4 2XR, United Kingdom
- * E-mail:
| |
Collapse
|
381
|
Hantel C, Beuschlein F. Xenograft models for adrenocortical carcinoma. Mol Cell Endocrinol 2016; 421:28-33. [PMID: 26033247 DOI: 10.1016/j.mce.2015.05.031] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Revised: 05/26/2015] [Accepted: 05/26/2015] [Indexed: 12/30/2022]
Abstract
Adrenocortical carcinomas (ACCs) are rare, heterogeneous and very malignant endocrine tumors with a poor prognosis. An important prerequisite to optimize existing therapeutic regimens and to develop novel therapeutic strategies are preclinical disease models. In recent years molecular and genetic profiling of surgical tumor specimen led to the identification of novel interesting markers. However, precise involvement of these markers in tumorigenesis and their functional relevance in therapeutic outcome is still under investigation. Xenograft models are important tools for such functional studies as they bear the potential to mimic the complexity of solid tumors including tumor cells, stroma and blood vessels. Thus, for the successful and safe development of novel therapeutic strategies xenograft models remain to be indispensable experimental tools. Here we provide an overview on currently existing xenograft models for ACC, their tissue origins, establishment, implications as well as limitations.
Collapse
Affiliation(s)
- Constanze Hantel
- Endocrine Research Unit, Medizinische Klinik und Poliklinik IV, Ludwig-Maximilians-Universität, Munich, Germany.
| | - Felix Beuschlein
- Endocrine Research Unit, Medizinische Klinik und Poliklinik IV, Ludwig-Maximilians-Universität, Munich, Germany
| |
Collapse
|
382
|
Kuracha MR, Thomas P, Loggie BW, Govindarajan V. Patient-derived xenograft mouse models of pseudomyxoma peritonei recapitulate the human inflammatory tumor microenvironment. Cancer Med 2016; 5:711-9. [PMID: 26833741 PMCID: PMC4831290 DOI: 10.1002/cam4.640] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Revised: 12/04/2015] [Accepted: 12/15/2015] [Indexed: 12/13/2022] Open
Abstract
Pseudomyxoma peritonei (PMP) is a neoplastic syndrome characterized by peritoneal tumor implants with copious mucinous ascites. The standard of care for PMP patients is aggressive cytoreductive surgery performed in conjunction with heated intraperitoneal chemotherapy. Not all patients are candidates for these procedures and a majority of the patients will have recurrent disease. In addition to secreted mucin, inflammation and fibrosis are central to PMP pathogenesis but the molecular processes that regulate tumor-stromal interactions within the peritoneal tumor microenvironment remain largely unknown. This knowledge is critical not only to elucidate PMP pathobiology but also to identify novel targets for therapy. Here, we report the generation of patient-derived xenograft (PDX) mouse models for PMP and assess the ability of these models to replicate the inflammatory peritoneal microenvironment of human PMP patients. PDX mouse models of low- and high-grade PMP were generated and were of a similar histopathology as human PMP. Cytokines previously shown to be elevated in human PMP were also elevated in PDX ascites. Significant differences in IL-6 and IL-8/KC/MIP2 were seen between human and PDX ascites. Interestingly, these cytokines were mostly secreted by mouse-derived, tumor-associated stromal cells rather than by human-derived PMP tumor cells. Our data suggest that the PMP PDX mouse models are especially suited to the study of tumor-stromal interactions that regulate the peritoneal inflammatory environment in PMP as the tumor and stromal cells in these mouse models are of human and murine origins, respectively. These mouse models are therefore, likely to be useful in vivo surrogates for testing and developing novel therapeutic treatment interventions for PMP.
Collapse
Affiliation(s)
- Murali R Kuracha
- Department of Surgery, Creighton University, 2500 California Plaza, Omaha, Nebraska, 68178
| | - Peter Thomas
- Department of Surgery, Creighton University, 2500 California Plaza, Omaha, Nebraska, 68178
| | - Brian W Loggie
- Department of Surgery, Creighton University, 2500 California Plaza, Omaha, Nebraska, 68178
| | - Venkatesh Govindarajan
- Department of Surgery, Creighton University, 2500 California Plaza, Omaha, Nebraska, 68178.,Department of Biomedical Sciences, Creighton University, 2500 California Plaza, Omaha, Nebraska, 68178
| |
Collapse
|
383
|
Animal models of colorectal cancer with liver metastasis. Cancer Lett 2016; 387:114-120. [PMID: 26850374 DOI: 10.1016/j.canlet.2016.01.048] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Revised: 01/26/2016] [Accepted: 01/26/2016] [Indexed: 01/01/2023]
Abstract
Liver metastasis is a leading cause of death in patients with colorectal cancer. Investigating the mechanisms of liver metastasis and control of disease progression are important strategies for improving survival of these patients. Liver metastasis is a multi-step process and relevant models representing these steps are necessary to understand the mechanism of liver metastasis and establish appropriate treatments. Recently, the development of animal models for use in metastasis research has greatly increased; however, there is still a lack of models that sufficiently represent human cancer. Thus, in order to select an optimal model for of a given study, it is necessary to fully understand the characteristics of each animal model. In this review, we describe the mouse models currently used for colorectal cancer with liver metastasis, their characteristics, and their pros and cons. This may help us specify the mechanism of liver metastasis and provide evidence relevant to clinical applications.
Collapse
|
384
|
Cho SY, Kang W, Han JY, Min S, Kang J, Lee A, Kwon JY, Lee C, Park H. An Integrative Approach to Precision Cancer Medicine Using Patient-Derived Xenografts. Mol Cells 2016; 39:77-86. [PMID: 26831452 PMCID: PMC4757806 DOI: 10.14348/molcells.2016.2350] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 12/23/2015] [Indexed: 12/16/2022] Open
Abstract
Cancer is a heterogeneous disease caused by diverse genomic alterations in oncogenes and tumor suppressor genes. Despite recent advances in high-throughput sequencing technologies and development of targeted therapies, novel cancer drug development is limited due to the high attrition rate from clinical studies. Patient-derived xenografts (PDX), which are established by the transfer of patient tumors into immunodeficient mice, serve as a platform for co-clinical trials by enabling the integration of clinical data, genomic profiles, and drug responsiveness data to determine precisely targeted therapies. PDX models retain many of the key characteristics of patients' tumors including histology, genomic signature, cellular heterogeneity, and drug responsiveness. These models can also be applied to the development of biomarkers for drug responsiveness and personalized drug selection. This review summarizes our current knowledge of this field, including methodologic aspects, applications in drug development, challenges and limitations, and utilization for precision cancer medicine.
Collapse
Affiliation(s)
- Sung-Yup Cho
- Department of Life Science, Ewha Womans University, Seoul 120-750,
Korea
| | - Wonyoung Kang
- Department of Life Science, Ewha Womans University, Seoul 120-750,
Korea
| | - Jee Yun Han
- Department of Life Science, Ewha Womans University, Seoul 120-750,
Korea
| | - Seoyeon Min
- Department of Life Science, Ewha Womans University, Seoul 120-750,
Korea
| | - Jinjoo Kang
- Department of Life Science, Ewha Womans University, Seoul 120-750,
Korea
| | - Ahra Lee
- Department of Life Science, Ewha Womans University, Seoul 120-750,
Korea
| | - Jee Young Kwon
- Department of Life Science, Ewha Womans University, Seoul 120-750,
Korea
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032,
USA
| | - Charles Lee
- Department of Life Science, Ewha Womans University, Seoul 120-750,
Korea
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032,
USA
| | - Hansoo Park
- Department of Life Science, Ewha Womans University, Seoul 120-750,
Korea
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032,
USA
| |
Collapse
|
385
|
Stone HB, Bernhard EJ, Coleman CN, Deye J, Capala J, Mitchell JB, Brown JM. Preclinical Data on Efficacy of 10 Drug-Radiation Combinations: Evaluations, Concerns, and Recommendations. Transl Oncol 2016; 9:46-56. [PMID: 26947881 PMCID: PMC4800059 DOI: 10.1016/j.tranon.2016.01.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Revised: 01/05/2016] [Accepted: 01/06/2016] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Clinical testing of new therapeutic interventions requires comprehensive, high-quality preclinical data. Concerns regarding quality of preclinical data have been raised in recent reports. This report examines the data on the interaction of 10 drugs with radiation and provides recommendations for improving the quality, reproducibility, and utility of future studies. The drugs were AZD6244, bortezomib, 17-DMAG, erlotinib, gefitinib, lapatinib, oxaliplatin/Lipoxal, sunitinib (Pfizer, Corporate headquarters, New York, NY), thalidomide, and vorinostat. METHODS In vitro and in vivo data were tabulated from 125 published papers, including methods, radiation and drug doses, schedules of administration, assays, measures of interaction, presentation and interpretation of data, dosimetry, and conclusions. RESULTS In many instances, the studies contained inadequate or unclear information that would hamper efforts to replicate or intercompare the studies, and that weakened the evidence for designing and conducting clinical trials. The published reports on these drugs showed mixed results on enhancement of radiation response, except for sunitinib, which was ineffective. CONCLUSIONS There is a need for improved experimental design, execution, and reporting of preclinical testing of agents that are candidates for clinical use in combination with radiation. A checklist is provided for authors and reviewers to ensure that preclinical studies of drug-radiation combinations meet standards of design, execution, and interpretation, and report necessary information to ensure high quality and reproducibility of studies. Improved design, execution, common measures of enhancement, and consistent interpretation of preclinical studies of drug-radiation interactions will provide rational guidance for prioritizing drugs for clinical radiotherapy trials and for the design of such trials.
Collapse
Affiliation(s)
- Helen B Stone
- Radiation Research Program, National Cancer Institute, 9609 Medical Center Dr, Rockville, 20850, MSC 9727
| | - Eric J Bernhard
- Radiation Research Program, National Cancer Institute, 9609 Medical Center Dr, Rockville, 20850, MSC 9727.
| | - C Norman Coleman
- Radiation Research Program, National Cancer Institute, 9609 Medical Center Dr, Rockville, 20850, MSC 9727
| | - James Deye
- Radiation Research Program, National Cancer Institute, 9609 Medical Center Dr, Rockville, 20850, MSC 9727
| | - Jacek Capala
- Radiation Research Program, National Cancer Institute, 9609 Medical Center Dr, Rockville, 20850, MSC 9727
| | - James B Mitchell
- Radiation Biology Branch, National Cancer Institute, MSC 1002, 10 Center Dr, Bethesda, MD, 20892
| | - J Martin Brown
- Stanford University, Radiation and Cancer Biology, CCSR-S Rm 1255, 269 Campus Dr, Stanford, CA, 94305
| |
Collapse
|
386
|
Guerrera F, Tabbò F, Bessone L, Maletta F, Gaudiano M, Ercole E, Annaratone L, Todaro M, Boita M, Filosso PL, Solidoro P, Delsedime L, Oliaro A, Sapino A, Ruffini E, Inghirami G. The Influence of Tissue Ischemia Time on RNA Integrity and Patient-Derived Xenografts (PDX) Engraftment Rate in a Non-Small Cell Lung Cancer (NSCLC) Biobank. PLoS One 2016; 11:e0145100. [PMID: 26731692 PMCID: PMC4701130 DOI: 10.1371/journal.pone.0145100] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2015] [Accepted: 11/28/2015] [Indexed: 01/13/2023] Open
Abstract
Introduction Bio-repositories are invaluable resources to implement translational cancer research and clinical programs. They represent one of the most powerful tools for biomolecular studies of clinically annotated cohorts, but high quality samples are required to generate reliable molecular readouts and functional studies. The objective of our study was to define the impact of cancer tissue ischemia time on RNA and DNA quality, and for the generation of Patient-Derived Xenografts (PDXs). Methods One-hundred thirty-five lung cancer specimens were selected among our Institutional BioBank samples. Associations between different warm (surgical) and cold (ex-vivo) ischemia time ranges and RNA quality or PDXs engraftment rates were assessed. RNA quality was determined by RNA integrity number (RINs) values. Fresh viable tissue fragments were implanted subcutaneously in NSG mice and serially transplanted. Results RNAs with a RIN>7 were detected in 51% of the sample (70/135), with values of RIN significantly lower (OR 0.08, P = 0.01) in samples preserved for more than 3 hours before cryopreservation. Higher quality DNA samples had a concomitant high RIN. Sixty-three primary tumors (41 adenocarcinoma) were implanted with an overall engraftment rate of 33%. Both prolonged warm (>2 hours) and ex-vivo ischemia time (>10 hours) were associated to a lower engraftment rate (OR 0.09 P = 0.01 and OR 0.04 P = 0.008, respectively). Conclusion RNA quality and PDXs engraftment rate were adversely affected by prolonged ischemia times. Proper tissue collection and processing reduce failure rate. Overall, NSCLC BioBanking represents an innovative modality, which can be successfully executed in routine clinical settings, when stringent Standard Operating Procedures are adopted.
Collapse
MESH Headings
- Aged
- Animals
- Carcinoma, Non-Small-Cell Lung/blood supply
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/pathology
- DNA, Neoplasm/genetics
- DNA, Neoplasm/metabolism
- Female
- Graft Survival
- Humans
- Interleukin Receptor Common gamma Subunit/deficiency
- Interleukin Receptor Common gamma Subunit/genetics
- Ischemia
- Lung Neoplasms/blood supply
- Lung Neoplasms/genetics
- Lung Neoplasms/pathology
- Male
- Mice, Inbred NOD
- Mice, Knockout
- Mice, SCID
- Middle Aged
- Multiplex Polymerase Chain Reaction
- RNA Stability
- RNA, Neoplasm/genetics
- RNA, Neoplasm/metabolism
- Retrospective Studies
- Reverse Transcriptase Polymerase Chain Reaction
- Time Factors
- Tissue Banks
- Transplantation, Heterologous
Collapse
Affiliation(s)
- Francesco Guerrera
- Department of Thoracic Surgery, University of Torino, 10126, Torino, Italy
- * E-mail:
| | - Fabrizio Tabbò
- Department of Molecular Biotechnology and Health Science and Center for Experimental Research and Medical Studies, University of Torino, 10126, Torino, Italy
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY, 10021, United States of America
| | - Luca Bessone
- Department of Molecular Biotechnology and Health Science and Center for Experimental Research and Medical Studies, University of Torino, 10126, Torino, Italy
| | - Francesca Maletta
- Department of Medical Sciences, University of Torino, 10126, Torino, Italy
| | - Marcello Gaudiano
- Department of Molecular Biotechnology and Health Science and Center for Experimental Research and Medical Studies, University of Torino, 10126, Torino, Italy
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY, 10021, United States of America
| | - Elisabetta Ercole
- Department of Molecular Biotechnology and Health Science and Center for Experimental Research and Medical Studies, University of Torino, 10126, Torino, Italy
| | - Laura Annaratone
- Department of Medical Sciences, University of Torino, 10126, Torino, Italy
| | - Maria Todaro
- Department of Molecular Biotechnology and Health Science and Center for Experimental Research and Medical Studies, University of Torino, 10126, Torino, Italy
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY, 10021, United States of America
| | - Monica Boita
- Department of Medical Sciences, Allergology and Immunology, University of Torino, 10126, Torino, Italy
| | - Pier Luigi Filosso
- Department of Thoracic Surgery, University of Torino, 10126, Torino, Italy
| | - Paolo Solidoro
- Unit of Pulmonology, San Giovanni Battista Hospital, 10126, Torino, Italy
| | - Luisa Delsedime
- Department of Medical Sciences, University of Torino, 10126, Torino, Italy
| | - Alberto Oliaro
- Department of Thoracic Surgery, University of Torino, 10126, Torino, Italy
| | - Anna Sapino
- Department of Medical Sciences, University of Torino, 10126, Torino, Italy
| | - Enrico Ruffini
- Department of Thoracic Surgery, University of Torino, 10126, Torino, Italy
| | - Giorgio Inghirami
- Department of Molecular Biotechnology and Health Science and Center for Experimental Research and Medical Studies, University of Torino, 10126, Torino, Italy
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY, 10021, United States of America
- Department of Pathology and NYU Cancer Center, New York University School of Medicine, New York, NY, 10016, United States of America
| |
Collapse
|
387
|
The Extraordinary Progress in Very Early Cancer Diagnosis and Personalized Therapy: The Role of Oncomarkers and Nanotechnology. JOURNAL OF NANOTECHNOLOGY 2016. [DOI: 10.1155/2016/3020361] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The impact of nanotechnology on oncology is revolutionizing cancer diagnosis and therapy and largely improving prognosis. This is mainly due to clinical translation of the most recent findings in cancer research, that is, the application of bio- and nanotechnologies. Cancer genomics and early diagnostics are increasingly playing a key role in developing more precise targeted therapies for most human tumors. In the last decade, accumulation of basic knowledge has resulted in a tremendous breakthrough in this field. Nanooncology, through the discovery of new genetic and epigenetic biomarkers, has facilitated the development of more sensitive biosensors for early cancer detection and cutting-edge multifunctionalized nanoparticles for tumor imaging and targeting. In the near future, nanooncology is expected to enable a very early tumor diagnosis, combined with personalized therapeutic approaches.
Collapse
|
388
|
Abstract
Gastric cancer is an important healthcare problem and represents the second leading cause of death for malignant disease worldwide. In the Western world, the diagnosis is done at late stage when treatments can be only palliative. Searches for new therapeutic regimens as well as for new biomarkers are in progress. To reduce cancer mortality is crucial the prevention of the lesion at earlier stages. Therefore, new bullets to prevention are needed. Nowadays, studies relating to different kinds of tumor are unanimous in considering cancer stem cells (CSCs) as "the core" of the tumor and the responsible of tumor chemoresistance and relapse. This chapter aims to provide the instructions to (1) isolate, (2) grow, and (3) validate, both in vivo and in vitro, the gastric CSC subpopulation.
Collapse
|
389
|
A Novel Immunocompetent Mouse Model of Pancreatic Cancer with Robust Stroma: a Valuable Tool for Preclinical Evaluation of New Therapies. J Gastrointest Surg 2016; 20:53-65; discussion 65. [PMID: 26582596 PMCID: PMC5724755 DOI: 10.1007/s11605-015-2985-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2015] [Accepted: 10/10/2015] [Indexed: 01/31/2023]
Abstract
A valid preclinical tumor model should recapitulate the tumor microenvironment. Immune and stromal components are absent in immunodeficient models of pancreatic cancer. While these components are present in genetically engineered models such as Kras(G12D); Trp53(R172H); Pdx-1Cre (KPC), immense variability in development of invasive disease makes them unsuitable for evaluation of novel therapies. We have generated a novel mouse model of pancreatic cancer by implanting tumor fragments from KPC mice into the pancreas of wild type mice. Three-millimeter tumor pieces from KPC mice were implanted into the pancreas of C57BL/6J mice. Four to eight weeks later, tumors were harvested, and stromal and immune components were evaluated. The efficacy of Minnelide, a novel compound which has been shown to be effective against pancreatic cancer in a number of preclinical murine models, was evaluated. In our model, consistent tumor growth and metastases were observed. Tumors demonstrated intense desmoplasia and leukocytic infiltration which was comparable to that in the genetically engineered KPC model and significantly more than that observed in KPC tumor-derived cell line implantation model. Minnelide treatment resulted in a significant decrease in the tumor weight and volume. This novel model demonstrates a consistent growth rate and tumor-associated mortality and recapitulates the tumor microenvironment. This convenient model is a valuable tool to evaluate novel therapies.
Collapse
|
390
|
van Marion DM, Domanska UM, Timmer-Bosscha H, Walenkamp AM. Studying cancer metastasis: Existing models, challenges and future perspectives. Crit Rev Oncol Hematol 2016; 97:107-17. [DOI: 10.1016/j.critrevonc.2015.08.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 08/05/2015] [Indexed: 02/03/2023] Open
|
391
|
Zhang C, Ren J, Yang Y, Wang D, He J, Huo D, Hu Y. Ultra-sensitive diagnosis of orthotopic patient derived hepatocellular carcinoma by Fe@graphene nanoparticles in MRI. RSC Adv 2016. [DOI: 10.1039/c6ra23511e] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
mPEG-DSPE modified Fe/graphene nanoparticles showed low cytotoxicity and high magnetic performance, providing super MRI diagnostic ability for cancer detection.
Collapse
Affiliation(s)
- Chao Zhang
- Institute of Materials Engineering
- College of Engineering and Applied Sciences
- Nanjing University
- P. R. China
- Department of Radiology
| | - Jing Ren
- The State Key Laboratory of Pharmaceutical Biotechnology
- Division of Immunology
- Medical School
- Nanjing University
- Nanjing 210093
| | - Yutong Yang
- Institute of Materials Engineering
- College of Engineering and Applied Sciences
- Nanjing University
- P. R. China
| | - Dunhui Wang
- Department of Physics
- Nanjing University
- Nanjing 210093
- P. R. China
| | - Jian He
- Department of Radiology
- Drum Tower Hospital
- School of Medicine
- Nanjing University
- P. R. China
| | - Da Huo
- Institute of Materials Engineering
- College of Engineering and Applied Sciences
- Nanjing University
- P. R. China
| | - Yong Hu
- Institute of Materials Engineering
- College of Engineering and Applied Sciences
- Nanjing University
- P. R. China
| |
Collapse
|
392
|
Kohli M, Wang L, Xie F, Sicotte H, Yin P, Dehm SM, Hart SN, Vedell PT, Barman P, Qin R, Mahoney DW, Carlson RE, Eckel-Passow JE, Atwell TD, Eiken PW, McMenomy BP, Wieben ED, Jha G, Jimenez RE, Weinshilboum R, Wang L. Mutational Landscapes of Sequential Prostate Metastases and Matched Patient Derived Xenografts during Enzalutamide Therapy. PLoS One 2015; 10:e0145176. [PMID: 26695660 PMCID: PMC4687867 DOI: 10.1371/journal.pone.0145176] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 11/30/2015] [Indexed: 12/22/2022] Open
Abstract
Developing patient derived models from individual tumors that capture the biological heterogeneity and mutation landscape in advanced prostate cancer is challenging, but essential for understanding tumor progression and delivery of personalized therapy in metastatic castrate resistant prostate cancer stage. To demonstrate the feasibility of developing patient derived xenograft models in this stage, we present a case study wherein xenografts were derived from cancer metastases in a patient progressing on androgen deprivation therapy and prior to initiating pre-chemotherapy enzalutamide treatment. Tissue biopsies from a metastatic rib lesion were obtained for sequencing before and after initiating enzalutamide treatment over a twelve-week period and also implanted subcutaneously as well as under the renal capsule in immuno-deficient mice. The genome and transcriptome landscapes of xenografts and the original patient tumor tissues were compared by performing whole exome and transcriptome sequencing of the metastatic tumor tissues and the xenografts at both time points. After comparing the somatic mutations, copy number variations, gene fusions and gene expression we found that the patient's genomic and transcriptomic alterations were preserved in the patient derived xenografts with high fidelity. These xenograft models provide an opportunity for predicting efficacy of existing and potentially novel drugs that is based on individual metastatic tumor expression signature and molecular pharmacology for delivery of precision medicine.
Collapse
Affiliation(s)
- Manish Kohli
- Department of Oncology, Mayo Clinic, Rochester, Minnesota, United States of America
- * E-mail: (MK); (Liguo Wang)
| | - Liguo Wang
- Division of Biomedical Statistics and Informatics, Department of Health Sciences, Mayo Clinic, Rochester, Minnesota, United States of America
- * E-mail: (MK); (Liguo Wang)
| | - Fang Xie
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Hugues Sicotte
- Division of Biomedical Statistics and Informatics, Department of Health Sciences, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Ping Yin
- Department of Oncology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Scott M. Dehm
- Masonic Cancer Center and Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Steven N. Hart
- Division of Biomedical Statistics and Informatics, Department of Health Sciences, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Peter T. Vedell
- Division of Biomedical Statistics and Informatics, Department of Health Sciences, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Poulami Barman
- Division of Biomedical Statistics and Informatics, Department of Health Sciences, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Rui Qin
- Division of Biomedical Statistics and Informatics, Department of Health Sciences, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Douglas W. Mahoney
- Division of Biomedical Statistics and Informatics, Department of Health Sciences, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Rachel E. Carlson
- Division of Biomedical Statistics and Informatics, Department of Health Sciences, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Jeanette E. Eckel-Passow
- Division of Biomedical Statistics and Informatics, Department of Health Sciences, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Thomas D. Atwell
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Patrick W. Eiken
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Brendan P. McMenomy
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Eric D. Wieben
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Gautam Jha
- Division of Hematology-Oncology, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Rafael E. Jimenez
- Department of Pathology and Lab Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Richard Weinshilboum
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Liewei Wang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota, United States of America
| |
Collapse
|
393
|
Li H, Wheeler S, Park Y, Ju Z, Thomas SM, Fichera M, Egloff AM, Lui VW, Duvvuri U, Bauman JE, Mills GB, Grandis JR. Proteomic Characterization of Head and Neck Cancer Patient-Derived Xenografts. Mol Cancer Res 2015; 14:278-86. [PMID: 26685214 DOI: 10.1158/1541-7786.mcr-15-0354] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 11/25/2015] [Indexed: 11/16/2022]
Abstract
UNLABELLED Despite advances in treatment approaches for head and neck squamous cell carcinoma (HNSCC), survival rates have remained stagnant due to the paucity of preclinical models that accurately reflect the human tumor. Patient-derived xenografts (PDX) are an emerging model system where patient tumors are implanted directly into mice. Increased understanding of the application and limitations of PDXs will facilitate their rational use. Studies to date have not reported protein profiles of PDXs. Therefore, we developed a large cohort of HNSCC PDXs and found that tumor take rate was not influenced by the clinical, pathologic, or processing features. Protein expression profiles, from a subset of the PDXs, were characterized by reverse-phase protein array and the data was compared with The Cancer Genome Atlas HNSCC data. Cluster analysis revealed that HNSCC PDXs were more similar to primary HNSCC than to any other tumor type. Interestingly, while a significant fraction of proteins were expressed similarly in both primary HNSCC and PDXs, a subset of proteins/phosphoproteins were expressed at higher (or lower) levels in PDXs compared with primary HNSCC. These findings indicate that the proteome is generally conserved in PDXs, but mechanisms for both positive and negative model selection and/or differences in the stromal components exist. IMPLICATIONS Proteomic characterization of HNSCC PDXs demonstrates potential drivers for model selection and provides a framework for improved utilization of this expanding model system.
Collapse
Affiliation(s)
- Hua Li
- Department of Otolaryngology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Sarah Wheeler
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Yongseok Park
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Zhenlin Ju
- Department of Bioinformatics and Computational Biology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sufi M Thomas
- Departments of Otolaryngology and Cancer Biology, Kansas University Medical Center, Kansas City, Kansas
| | - Michele Fichera
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Ann M Egloff
- Department of Otolaryngology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Vivian W Lui
- Department of Pharmacology and Pharmacy, University of Hong Kong, Hong Kong
| | - Umamaheswar Duvvuri
- Department of Otolaryngology, University of Pittsburgh, Pittsburgh, Pennsylvania. Veterans Affairs Pittsburgh Healthcare System, University Drive Campus, Pittsburgh, Pennsylvania
| | - Julie E Bauman
- Department of Internal Medicine - Hematology/Oncology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Gordon B Mills
- Department of Bioinformatics and Computational Biology, University of Texas MD Anderson Cancer Center, Houston, Texas. Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jennifer R Grandis
- Department of Otolaryngology, University of Pittsburgh, Pittsburgh, Pennsylvania. Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania. Department of Otolaryngology-Head and Neck Surgery, University of California San Francisco, San Francisco, California.
| |
Collapse
|
394
|
Loizides C, Iacovides D, Hadjiandreou MM, Rizki G, Achilleos A, Strati K, Mitsis GD. Model-Based Tumor Growth Dynamics and Therapy Response in a Mouse Model of De Novo Carcinogenesis. PLoS One 2015; 10:e0143840. [PMID: 26649886 PMCID: PMC4674149 DOI: 10.1371/journal.pone.0143840] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Accepted: 11/10/2015] [Indexed: 12/17/2022] Open
Abstract
Tumorigenesis is a complex, multistep process that depends on numerous alterations within the cell and contribution from the surrounding stroma. The ability to model macroscopic tumor evolution with high fidelity may contribute to better predictive tools for designing tumor therapy in the clinic. However, attempts to model tumor growth have mainly been developed and validated using data from xenograft mouse models, which fail to capture important aspects of tumorigenesis including tumor-initiating events and interactions with the immune system. In the present study, we investigate tumor growth and therapy dynamics in a mouse model of de novo carcinogenesis that closely recapitulates tumor initiation, progression and maintenance in vivo. We show that the rate of tumor growth and the effects of therapy are highly variable and mouse specific using a Gompertz model to describe tumor growth and a two-compartment pharmacokinetic/ pharmacodynamic model to describe the effects of therapy in mice treated with 5-FU. We show that inter-mouse growth variability is considerably larger than intra-mouse variability and that there is a correlation between tumor growth and drug kill rates. Our results show that in vivo tumor growth and regression in a double transgenic mouse model are highly variable both within and between subjects and that mathematical models can be used to capture the overall characteristics of this variability. In order for these models to become useful tools in the design of optimal therapy strategies and ultimately in clinical practice, a subject-specific modelling strategy is necessary, rather than approaches that are based on the average behavior of a given subject population which could provide erroneous results.
Collapse
Affiliation(s)
- Charalambos Loizides
- Department of Electrical & Electronic Engineering & KIOS Research Center for Intelligent Systems & Networks, University of Cyprus, Nicosia, Cyprus
| | - Demetris Iacovides
- Department of Biological Sciences, University of Cyprus, Nicosia, Cyprus
| | - Marios M. Hadjiandreou
- Department of Electrical & Electronic Engineering & KIOS Research Center for Intelligent Systems & Networks, University of Cyprus, Nicosia, Cyprus
| | - Gizem Rizki
- Department of Biological Sciences, University of Cyprus, Nicosia, Cyprus
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, United States of America
| | - Achilleas Achilleos
- Department of Electrical & Electronic Engineering & KIOS Research Center for Intelligent Systems & Networks, University of Cyprus, Nicosia, Cyprus
| | - Katerina Strati
- Department of Biological Sciences, University of Cyprus, Nicosia, Cyprus
- * E-mail: (KS); (GM)
| | - Georgios D. Mitsis
- Department of Electrical & Electronic Engineering & KIOS Research Center for Intelligent Systems & Networks, University of Cyprus, Nicosia, Cyprus
- Department of Bioengineering, McGill University, Montreal QC, Canada
- * E-mail: (KS); (GM)
| |
Collapse
|
395
|
Grimm D, Zolotukhin S. E Pluribus Unum: 50 Years of Research, Millions of Viruses, and One Goal--Tailored Acceleration of AAV Evolution. Mol Ther 2015; 23:1819-31. [PMID: 26388463 PMCID: PMC4700111 DOI: 10.1038/mt.2015.173] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 09/10/2015] [Indexed: 12/11/2022] Open
Abstract
Fifty years ago, a Science paper by Atchison et al. reported a newly discovered virus that would soon become known as adeno-associated virus (AAV) and that would subsequently emerge as one of the most versatile and most auspicious vectors for human gene therapy. A large part of its attraction stems from the ease with which the viral capsid can be engineered for particle retargeting to cell types of choice, evasion from neutralizing antibodies or other desirable properties. Particularly powerful and in the focus of the current review are high-throughput methods aimed at expanding the repertoire of AAV vectors by means of directed molecular evolution, such as random mutagenesis, DNA family shuffling, in silico reconstruction of ancestral capsids, or peptide display. Here, unlike the wealth of prior reviews on this topic, we especially emphasize and critically discuss the practical aspects of the different procedures that affect the ultimate outcome, including diversification protocols, combinatorial library complexity, and selection strategies. Our overall aim is to provide general guidance that should help users at any level, from novice to expert, to safely navigate through the rugged space of directed AAV evolution while avoiding the pitfalls that are associated with these challenging but promising technologies.
Collapse
Affiliation(s)
- Dirk Grimm
- Department of Infectious Diseases/Virology, Cluster of Excellence CellNetworks, Heidelberg University Hospital, Heidelberg, Germany
| | - Sergei Zolotukhin
- Division of Cell and Molecular Therapy, Department of Pediatrics, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
396
|
Friedman AA, Letai A, Fisher DE, Flaherty KT. Precision medicine for cancer with next-generation functional diagnostics. Nat Rev Cancer 2015; 15:747-56. [PMID: 26536825 PMCID: PMC4970460 DOI: 10.1038/nrc4015] [Citation(s) in RCA: 405] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Precision medicine is about matching the right drugs to the right patients. Although this approach is technology agnostic, in cancer there is a tendency to make precision medicine synonymous with genomics. However, genome-based cancer therapeutic matching is limited by incomplete biological understanding of the relationship between phenotype and cancer genotype. This limitation can be addressed by functional testing of live patient tumour cells exposed to potential therapies. Recently, several 'next-generation' functional diagnostic technologies have been reported, including novel methods for tumour manipulation, molecularly precise assays of tumour responses and device-based in situ approaches; these address the limitations of the older generation of chemosensitivity tests. The promise of these new technologies suggests a future diagnostic strategy that integrates functional testing with next-generation sequencing and immunoprofiling to precisely match combination therapies to individual cancer patients.
Collapse
Affiliation(s)
- Adam A Friedman
- Massachusetts General Hospital Cancer Center, Harvard Medical School, 55 Fruit Street, Boston, Massachusetts 02114, USA
| | - Anthony Letai
- Dana-Farber Cancer Institute, Harvard Medical School, 440 Brookline Avenue, Mayer 430, Boston, Massachusetts 02215, USA
| | - David E Fisher
- Massachusetts General Hospital Cancer Center, Harvard Medical School, 55 Fruit Street, Boston, Massachusetts 02114, USA
- Dermatology and Cutaneous Biology Research Center, Massachusetts General Hospital, 149 East 13th Street, Charlestown, Massachusetts 02129, USA
| | - Keith T Flaherty
- Massachusetts General Hospital Cancer Center, Harvard Medical School, 55 Fruit Street, Boston, Massachusetts 02114, USA
| |
Collapse
|
397
|
Jiang Y, Wu Q, Yang X, Zhao J, Jin Y, Li K, Ma Y, Chen X, Tian F, Zhao S, Xu J, Lu J, Yin X, Liu K, Dong Z. A method for establishing a patient-derived xenograft model to explore new therapeutic strategies for esophageal squamous cell carcinoma. Oncol Rep 2015; 35:785-92. [PMID: 26718633 DOI: 10.3892/or.2015.4459] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2015] [Accepted: 10/10/2015] [Indexed: 11/05/2022] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is the predominant histological type of esophageal carcinoma in China. The overall 5-year survival rate of ESCC patients is in the low range of 15-25%. One important reason for the poor prognosis is that the underlying molecular mechanisms are unclear. Furthermore, the development of effective therapeutic strategies to improve patient outcome is needed. Animal models can be beneficial to analyze the molecular mechanisms as well as specific clinical therapeutic strategies for esophageal cancer. In recent years, patient-derived xenografts (PDXs) have been widely used in numerous types of cancers to investigate the basic mechanisms and to conduct preclinical research. Accumulating evidence indicates that the PDX model is an important tool for basic and clinical research. Herein, we successfully established 14 ESCC PDXs. These PDX models preserved the patient pathological characteristics and effectively reflected the patient biological heterogeneity. Cancers exhibit diverse growth rates and tumor texture, even more, they have different signaling pathways. The PDX model is a superior strategy for understanding the underlying molecular mechanisms of ESCC and for screening new therapeutic strategies for ESCC patients.
Collapse
Affiliation(s)
- Yanan Jiang
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Qiong Wu
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Xiawen Yang
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Jimin Zhao
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Yuxi Jin
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Ke Li
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Yihui Ma
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Xinhuan Chen
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Fang Tian
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Song Zhao
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Jinglong Xu
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Jing Lu
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Xueshan Yin
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Kangdong Liu
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Ziming Dong
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| |
Collapse
|
398
|
Alkema NG, Wisman GBA, van der Zee AGJ, van Vugt MATM, de Jong S. Studying platinum sensitivity and resistance in high-grade serous ovarian cancer: Different models for different questions. Drug Resist Updat 2015; 24:55-69. [PMID: 26830315 DOI: 10.1016/j.drup.2015.11.005] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Revised: 11/04/2015] [Accepted: 11/19/2015] [Indexed: 12/21/2022]
Abstract
High-grade serous ovarian cancer (HGSOC) has the highest mortality rate among all gynecological cancers. Patients are generally diagnosed in an advanced stage with the majority of cases displaying platinum resistant relapses. Recent genomic interrogation of large numbers of HGSOC patient samples indicated high complexity in terms of genetic aberrations, intra- and intertumor heterogeneity and underscored their lack of targetable oncogenic mutations. Sub-classifications of HGSOC based on expression profiles, termed 'differentiated', 'immunoreactive', 'mesenchymal' and 'proliferative', were shown to have prognostic value. In addition, in almost half of all HGSOC patients, a deficiency in homologous recombination (HR) was found that potentially can be targeted using PARP inhibitors. Developing precision medicine requires advanced experimental models. In the current review, we discuss experimental HGSOC models in which resistance to platinum therapy and the use of novel therapeutics can be carefully studied. Panels of better-defined primary cell lines need to be established to capture the full spectrum of HGSOC subtypes. Further refinement of cell lines is obtained with a 3-dimensional culture model mimicking the tumor microenvironment. Alternatively, ex vivo ovarian tumor tissue slices are used. For in vivo studies, larger panels of ovarian cancer patient-derived xenografts (PDXs) are being established, encompassing all expression subtypes. Ovarian cancer PDXs grossly retain tumor heterogeneity and clinical response to platinum therapy is preserved. PDXs are currently used in drug screens and as avatars for patient response. The role of the immune system in tumor responses can be assessed using humanized PDXs and immunocompetent genetically engineered mouse models. Dynamic tracking of genetic alterations in PDXs as well as patients during treatment and after relapse is feasible by sequencing circulating cell-free tumor DNA and analyzing circulating tumor cells. We discuss how various models and methods can be combined to delineate the molecular mechanisms underlying platinum resistance and to select HGSOC patients other than BRCA1/2-mutation carriers that could potentially benefit from the synthetic lethality of PARP inhibitors. This integrated approach is a first step to improve therapy outcomes in specific subgroups of HGSOC patients.
Collapse
Affiliation(s)
- Nicolette G Alkema
- Department of Gynecologic Oncology, Cancer Research Centre Groningen, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - G Bea A Wisman
- Department of Gynecologic Oncology, Cancer Research Centre Groningen, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Ate G J van der Zee
- Department of Gynecologic Oncology, Cancer Research Centre Groningen, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Marcel A T M van Vugt
- Department of Medical Oncology, Cancer Research Centre Groningen, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Steven de Jong
- Department of Medical Oncology, Cancer Research Centre Groningen, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| |
Collapse
|
399
|
Lau V, Wong ALA, Ng C, Mok Y, Lakshmanan M, Yan B. Drug sensitivity testing platforms for gastric cancer diagnostics. J Clin Pathol 2015; 69:93-6. [PMID: 26567318 DOI: 10.1136/jclinpath-2015-203426] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 10/20/2015] [Indexed: 01/09/2023]
Abstract
Gastric cancer diagnostics has traditionally been histomorphological and primarily the domain of surgical pathologists. Although there is an increasing usage of molecular and genomic techniques for clinical diagnostics, there is an emerging field of personalised drug sensitivity testing. In this review, we describe the various personalised drug sensitivity testing platforms and discuss the challenges facing clinical adoption of these assays for gastric cancer.
Collapse
Affiliation(s)
- Vianne Lau
- Department of Gastroenterology Monash University, Melbourne, Australia
| | - Andrea Li-Ann Wong
- Department of Haematology and Oncology, National University Health System, Singapore, Singapore
| | - Christopher Ng
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore Department of Laboratory Medicine, Molecular Diagnosis Centre, National University Hospital, National University Health System, Singapore, Singapore
| | - Yingting Mok
- Department of Pathology, National University Hospital, National University Health System, Singapore, Singapore
| | - Manikandan Lakshmanan
- Mouse Models for Human Cancer Unit, Institute of Molecular and Cell Biology, Singapore, Singapore
| | - Benedict Yan
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore Department of Laboratory Medicine, Molecular Diagnosis Centre, National University Hospital, National University Health System, Singapore, Singapore
| |
Collapse
|
400
|
Xie T, Musteanu M, Lopez-Casas PP, Shields DJ, Olson P, Rejto PA, Hidalgo M. Whole Exome Sequencing of Rapid Autopsy Tumors and Xenograft Models Reveals Possible Driver Mutations Underlying Tumor Progression. PLoS One 2015; 10:e0142631. [PMID: 26555578 PMCID: PMC4640827 DOI: 10.1371/journal.pone.0142631] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 10/23/2015] [Indexed: 12/31/2022] Open
Abstract
Pancreatic Ductal Adenocarcinoma (PDAC) is a highly lethal malignancy due to its propensity to invade and rapidly metastasize and remains very difficult to manage clinically. One major hindrance towards a better understanding of PDAC is the lack of molecular data sets and models representative of end stage disease. Moreover, it remains unclear how molecularly similar patient-derived xenograft (PDX) models are to the primary tumor from which they were derived. To identify potential molecular drivers in metastatic pancreatic cancer progression, we obtained matched primary tumor, metastases and normal (peripheral blood) samples under a rapid autopsy program and performed whole exome sequencing (WES) on tumor as well as normal samples. PDX models were also generated, sequenced and compared to tumors. Across the matched data sets generated for three patients, there were on average approximately 160 single-nucleotide mutations in each sample. The majority of mutations in each patient were shared among the primary and metastatic samples and, importantly, were largely retained in the xenograft models. Based on the mutation prevalence in the primary and metastatic sites, we proposed possible clonal evolution patterns marked by functional mutations affecting cancer genes such as KRAS, TP53 and SMAD4 that may play an important role in tumor initiation, progression and metastasis. These results add to our understanding of pancreatic tumor biology, and demonstrate that PDX models derived from advanced or end-stage likely closely approximate the genetics of the disease in the clinic and thus represent a biologically and clinically relevant pre-clinical platform that may enable the development of effective targeted therapies for PDAC.
Collapse
Affiliation(s)
- Tao Xie
- Oncology Research, Pfizer Worldwide Research and Development, San Diego, California, United States of America
- * E-mail: (TX); (MH)
| | - Monica Musteanu
- CNIO (Spanish National Cancer Research Centre), E-28029, Madrid, Madrid, Spain
| | | | - David J. Shields
- Oncology Research, Pfizer Worldwide Research and Development, San Diego, California, United States of America
| | - Peter Olson
- Oncology Research, Pfizer Worldwide Research and Development, San Diego, California, United States of America
| | - Paul A. Rejto
- Oncology Research, Pfizer Worldwide Research and Development, San Diego, California, United States of America
| | - Manuel Hidalgo
- CNIO (Spanish National Cancer Research Centre), E-28029, Madrid, Madrid, Spain
- * E-mail: (TX); (MH)
| |
Collapse
|