1
|
Ishii T, Mimura I, Nagaoka K, Naito A, Sugasawa T, Kuroda R, Yamada D, Kanki Y, Kume H, Ushiku T, Kakimi K, Tanaka T, Nangaku M. Effect of M2-like macrophages of the injured-kidney cortex on kidney cancer progression. Cell Death Dis 2022; 8:480. [PMID: 36470862 PMCID: PMC9722672 DOI: 10.1038/s41420-022-01255-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 11/11/2022] [Accepted: 11/14/2022] [Indexed: 12/12/2022]
Abstract
Chronic kidney disease (CKD) affects kidney cancer patients' mortality. However, the underlying mechanism remains unknown. M2-like macrophages have pro-tumor functions, also exist in injured kidney, and promote kidney fibrosis. Thus, it is suspected that M2-like macrophages in injured kidney induce the pro-tumor microenvironment leading to kidney cancer progression. We found that M2-like macrophages present in the injured kidney promoted kidney cancer progression and induced resistance to anti-PD1 antibody through its pro-tumor function and inhibition of CD8+ T cell infiltration. RNA-seq revealed Slc7a11 was upregulated in M2-like macrophages. Inhibition of Slc7a11 with sulfasalazine inhibited the pro-tumor function of M2-like macrophages and synergized with anti-PD1 antibody. Moreover, SLC7A11-positive macrophages were associated with poor prognosis among kidney cancer patients. Collectively, this study dissects the characteristic microenvironment in the injured kidney that contributed to kidney cancer progression and anti-PD1 antibody resistance. This insight offers promising combination therapy with anti-PD1 antibody and macrophage targeted therapy.
Collapse
Affiliation(s)
- Taisuke Ishii
- grid.26999.3d0000 0001 2151 536XDivision of Nephrology and Endocrinology, The University of Tokyo Graduate School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 1138655 Japan
| | - Imari Mimura
- grid.26999.3d0000 0001 2151 536XDivision of Nephrology and Endocrinology, The University of Tokyo Graduate School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 1138655 Japan
| | - Koji Nagaoka
- grid.412708.80000 0004 1764 7572Department of Immunotherapeutics, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 1138655 Japan
| | - Akihiro Naito
- grid.26999.3d0000 0001 2151 536XDivision of Urology, The University of Tokyo Graduate School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 1138655 Japan
| | - Takehito Sugasawa
- grid.20515.330000 0001 2369 4728Laboratory of Clinical Examination/Sports Medicine, Division of Clinical Medicine, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 3058577 Japan
| | - Ryohei Kuroda
- grid.26999.3d0000 0001 2151 536XDepartment of Pathology, The University of Tokyo Graduate School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 1138655 Japan
| | - Daisuke Yamada
- grid.26999.3d0000 0001 2151 536XDivision of Urology, The University of Tokyo Graduate School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 1138655 Japan
| | - Yasuharu Kanki
- grid.20515.330000 0001 2369 4728Laboratory of Clinical Examination/Sports Medicine, Division of Clinical Medicine, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 3058577 Japan
| | - Haruki Kume
- grid.26999.3d0000 0001 2151 536XDivision of Urology, The University of Tokyo Graduate School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 1138655 Japan
| | - Tetsuo Ushiku
- grid.26999.3d0000 0001 2151 536XDepartment of Pathology, The University of Tokyo Graduate School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 1138655 Japan
| | - Kazuhiro Kakimi
- grid.412708.80000 0004 1764 7572Department of Immunotherapeutics, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 1138655 Japan
| | - Tetsuhiro Tanaka
- grid.26999.3d0000 0001 2151 536XDivision of Nephrology and Endocrinology, The University of Tokyo Graduate School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 1138655 Japan ,grid.69566.3a0000 0001 2248 6943Department of Nephrology, Rheumatology and Endocrinology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, 9808574 Japan
| | - Masaomi Nangaku
- grid.26999.3d0000 0001 2151 536XDivision of Nephrology and Endocrinology, The University of Tokyo Graduate School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 1138655 Japan
| |
Collapse
|
2
|
Lee HW, Seo HS, Yeom SY, Kim SN, Kim CR, Park DH, Park W, Choy YB, Park CG, Seo SI. Cabozantinib-Loaded PLGA Nanoparticles: A Potential Adjuvant Strategy for Surgically Resected High-Risk Non-Metastatic Renal Cell Carcinoma. Int J Mol Sci 2022; 23:12634. [PMID: 36293494 PMCID: PMC9604013 DOI: 10.3390/ijms232012634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 10/13/2022] [Accepted: 10/14/2022] [Indexed: 11/16/2022] Open
Abstract
Patients with high-risk non-metastatic renal cell carcinoma (RCC) are at risk of metastatic relapse following nephrectomy. Cabozantinib (CZ), a potent multitarget tyrosine kinase inhibitor, interferes with angiogenesis and immunosuppression associated with surgery-induced metastasis. Here, we explored the therapeutic potential of CZ-loaded poly(lactic-co-glycolic acid) (PLGA) nanoparticles (CZ-PLGA-NPs) as an adjuvant strategy for targeting post-nephrectomy metastasis. A clinically relevant subline recapitulating post-nephrectomy lung metastasis of high-risk human RCC, namely Renca-SRLu5-Luc, was established through in vivo serial selection of luciferase-expressing murine RCC Renca-Luc cells. CZ was encapsulated into PLGA-NPs via the conventional single emulsion technique. The multifaceted preclinical antimetastatic efficacy of CZ-PLGA-NPs was assessed in Renca-SRLu5-Luc cells. CZ-PLGA-NPs with a smooth surface displayed desirable physicochemical properties, good CZ encapsulation efficiency, as well as controlled and sustained CZ release. CZ-PLGA-NPs exhibited remarkable dose-dependent toxicity against Renca-SRLu5-Luc cells by inducing G2/M cell cycle arrest and apoptosis. CZ-PLGA-NPs attenuated in vitro colony formation, migration, and invasion by abrogating AKT and ERK1/2 activation. An intravenous injection of CZ-PLGA-NPs markedly reduced lung metastatic burden and prolonged lifespan with favorable safety in the Renca-SRLu5-Luc experimental lung metastasis model. The novel CZ-PLGA-NPs system with multifaceted antimetastatic effects and alleviating off-target toxicity potential is a promising adjunctive agent for patients with surgically resected high-risk RCC.
Collapse
Affiliation(s)
- Hye Won Lee
- Department of Urology, Center for Urologic Cancer, National Cancer Center, Goyang 10408, Korea
| | - Hee Seung Seo
- Department of Biomedical Engineering, SKKU Institute for Convergence, Sungkyunkwan University, Suwon 16419, Korea
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon 16419, Korea
| | - Seon-Yong Yeom
- Department of Urology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
| | - Se-Na Kim
- Institute of Medical & Biological Engineering, Medical Research Center, Seoul National University, Seoul 03080, Korea
| | - Cho Rim Kim
- Interdisciplinary Program in Bioengineering, College of Engineering, Seoul National University, Seoul 08826, Korea
| | - Dae-Hwan Park
- Department of Engineering Chemistry, College of Engineering, Chungbuk National University, Cheongju 28644, Korea
- Department of Industrial Cosmetic Science and Department of Synchrotron Radiation Science and Technology, College of Bio-Health University System, Chungbuk National University, Cheongju 28644, Korea
| | - Wooram Park
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon 16419, Korea
| | - Young Bin Choy
- Institute of Medical & Biological Engineering, Medical Research Center, Seoul National University, Seoul 03080, Korea
- Interdisciplinary Program in Bioengineering, College of Engineering, Seoul National University, Seoul 08826, Korea
- Department of Biomedical Engineering, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Chun Gwon Park
- Department of Biomedical Engineering, SKKU Institute for Convergence, Sungkyunkwan University, Suwon 16419, Korea
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon 16419, Korea
- Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Korea
| | - Seong Il Seo
- Department of Urology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
| |
Collapse
|
3
|
He X, Zhou S, Dolan M, Shi Y, Wang J, Quinn B, Jahagirdar D, Huang WC, Tsuji M, Pili R, Ito F, Ortega J, Abrams SI, Ebos JML, Lovell JF. Immunization with short peptide particles reveals a functional CD8 + T-cell neoepitope in a murine renal carcinoma model. J Immunother Cancer 2021; 9:jitc-2021-003101. [PMID: 34862254 PMCID: PMC8647534 DOI: 10.1136/jitc-2021-003101] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/04/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Induction of CD8+ T cells that recognize immunogenic, mutated protein fragments in the context of major histocompatibility class I (MHC-I) is a pressing challenge for cancer vaccine development. METHODS Using the commonly used murine renal adenocarcinoma RENCA cancer model, MHC-I restricted neoepitopes are predicted following next-generation sequencing. Candidate neoepitopes are screened in mice using a potent cancer vaccine adjuvant system that converts short peptides into immunogenic nanoparticles. An identified functional neoepitope vaccine is then tested in various therapeutic experimental tumor settings. RESULTS Conversion of 20 short MHC-I restricted neoepitope candidates into immunogenic nanoparticles results in antitumor responses with multivalent vaccination. Only a single neoepitope candidate, Nesprin-2 L4492R (Nes2LR), induced functional responses but still did so when included within 20-plex or 60-plex particles. Immunization with the short Nes2LR neoepitope with the immunogenic particle-inducing vaccine adjuvant prevented tumor growth at doses multiple orders of magnitude less than with other vaccine adjuvants, which were ineffective. Nes2LR vaccination inhibited or eradicated disease in subcutaneous, experimental lung metastasis and orthotopic tumor models, synergizing with immune checkpoint blockade. CONCLUSION These findings establish the feasibility of using short, MHC-I-restricted neoepitopes for straightforward immunization with multivalent or validated neoepitopes to induce cytotoxic CD8+ T cells. Furthermore, the Nes2LR neoepitope could be useful for preclinical studies involving renal cell carcinoma immunotherapy.
Collapse
Affiliation(s)
- Xuedan He
- Department of Biomedical Engineering, University at Buffalo, Buffalo, New York, USA
| | - Shiqi Zhou
- Department of Biomedical Engineering, University at Buffalo, Buffalo, New York, USA
| | - Melissa Dolan
- Department of Experimental Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Yuhao Shi
- Department of Experimental Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Jianxin Wang
- Center for Computational Research, University at Buffalo, Buffalo, NY, USA
| | - Breandan Quinn
- Department of Biomedical Engineering, University at Buffalo, Buffalo, New York, USA
| | - Dushyant Jahagirdar
- Department of Anatomy and Cell Biology, McGill University, Montreal, Québec, Canada
| | - Wei-Chiao Huang
- Department of Biomedical Engineering, University at Buffalo, Buffalo, New York, USA
| | - Moriya Tsuji
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Roberto Pili
- Department of Medicine, State University of New York, Buffalo, NY, USA
| | - Fumito Ito
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Joaquin Ortega
- Department of Anatomy and Cell Biology, McGill University, Montreal, Québec, Canada
| | - Scott I Abrams
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - John M L Ebos
- Department of Experimental Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Jonathan F Lovell
- Department of Biomedical Engineering, University at Buffalo, Buffalo, New York, USA
| |
Collapse
|
4
|
Glennon KI, Maralani M, Abdian N, Paccard A, Montermini L, Nam AJ, Arseneault M, Staffa A, Jandaghi P, Meehan B, Brimo F, Tanguay S, Rak J, Riazalhosseini Y. Rational Development of Liquid Biopsy Analysis in Renal Cell Carcinoma. Cancers (Basel) 2021; 13:cancers13225825. [PMID: 34830979 PMCID: PMC8616270 DOI: 10.3390/cancers13225825] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 11/15/2021] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Among patients affected by renal cell carcinoma (RCC), the most common type of kidney cancer, it remains difficult to identify those who are at high risk for relapse or metastasis. This is in part due to the absence of reliable clinical biomarkers and robust methods to capture them. The aim of our study was to develop an improved assay to capture prognostic genomic biomarkers in circulating tumor DNA (ctDNA) in RCC. For this purpose, we first established a next generation sequencing (NGS) assay, targeting genes that are tailored for RCC and that are largely excluded from commercially available assays. Next, we showed the reliable performance of this assay to detect prognostic gene mutations in tumor DNA isolated from plasma, and from extracellular vesicles. Thus, our study provides a resource to facilitate ctDNA analysis for precision medicine in RCC. Abstract Renal cell carcinoma (RCC) is known for its variable clinical behavior and outcome, including heterogeneity in developing relapse or metastasis. Recent data highlighted the potential of somatic mutations as promising biomarkers for risk stratification in RCC. Likewise, the analysis of circulating tumor DNA (ctDNA) for such informative somatic mutations (liquid biopsy) is considered an important advance for precision oncology in RCC, allowing to monitor molecular disease evolution in real time. However, our knowledge about the utility of ctDNA analysis in RCC is limited, in part due to the lack of RCC-appropriate assays for ctDNA analysis. Here, by interrogating different blood compartments in xenograft models, we identified plasma cell-free (cf) DNA and extracellular vesicles (ev) DNA enriched for RCC-associated ctDNA. Additionally, we developed sensitive targeted sequencing and bioinformatics workflows capable of detecting somatic mutations in RCC-relevant genes with allele frequencies ≥ 0.5%. Applying this assay to patient-matched tumor and liquid biopsies, we captured tumor mutations in cf- and ev-DNA fractions isolated from the blood, highlighting the potentials of both fractions for ctDNA analysis. Overall, our study presents an RCC-appropriate sequencing assay and workflow for ctDNA analysis and provides a proof of principle as to the feasibility of detecting tumor-specific mutations in liquid biopsy in RCC patients.
Collapse
Affiliation(s)
- Kate I. Glennon
- McGill Genome Centre, McGill University, 740 Doctor Penfield Avenue, Montreal, QC H3A 0G1, Canada; (K.I.G.); (M.M.); (A.P.); (A.J.N.); (M.A.); (A.S.); (P.J.)
- Department of Human Genetics, McGill University, 1205 Doctor Penfield Avenue, Montreal, QC H3A 1B1, Canada
| | - Mahafarin Maralani
- McGill Genome Centre, McGill University, 740 Doctor Penfield Avenue, Montreal, QC H3A 0G1, Canada; (K.I.G.); (M.M.); (A.P.); (A.J.N.); (M.A.); (A.S.); (P.J.)
- Department of Human Genetics, McGill University, 1205 Doctor Penfield Avenue, Montreal, QC H3A 1B1, Canada
- The Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada; (N.A.); (L.M.); (B.M.); (J.R.)
| | - Narges Abdian
- The Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada; (N.A.); (L.M.); (B.M.); (J.R.)
| | - Antoine Paccard
- McGill Genome Centre, McGill University, 740 Doctor Penfield Avenue, Montreal, QC H3A 0G1, Canada; (K.I.G.); (M.M.); (A.P.); (A.J.N.); (M.A.); (A.S.); (P.J.)
| | - Laura Montermini
- The Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada; (N.A.); (L.M.); (B.M.); (J.R.)
| | - Alice Jisoo Nam
- McGill Genome Centre, McGill University, 740 Doctor Penfield Avenue, Montreal, QC H3A 0G1, Canada; (K.I.G.); (M.M.); (A.P.); (A.J.N.); (M.A.); (A.S.); (P.J.)
| | - Madeleine Arseneault
- McGill Genome Centre, McGill University, 740 Doctor Penfield Avenue, Montreal, QC H3A 0G1, Canada; (K.I.G.); (M.M.); (A.P.); (A.J.N.); (M.A.); (A.S.); (P.J.)
| | - Alfredo Staffa
- McGill Genome Centre, McGill University, 740 Doctor Penfield Avenue, Montreal, QC H3A 0G1, Canada; (K.I.G.); (M.M.); (A.P.); (A.J.N.); (M.A.); (A.S.); (P.J.)
| | - Pouria Jandaghi
- McGill Genome Centre, McGill University, 740 Doctor Penfield Avenue, Montreal, QC H3A 0G1, Canada; (K.I.G.); (M.M.); (A.P.); (A.J.N.); (M.A.); (A.S.); (P.J.)
- Department of Human Genetics, McGill University, 1205 Doctor Penfield Avenue, Montreal, QC H3A 1B1, Canada
| | - Brian Meehan
- The Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada; (N.A.); (L.M.); (B.M.); (J.R.)
| | - Fadi Brimo
- Department of Pathology, McGill University, Montreal, QC H3A 2B4, Canada;
| | - Simon Tanguay
- Division of Urology, McGill University, Montreal, QC H4A 3J1, Canada;
| | - Janusz Rak
- The Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada; (N.A.); (L.M.); (B.M.); (J.R.)
| | - Yasser Riazalhosseini
- McGill Genome Centre, McGill University, 740 Doctor Penfield Avenue, Montreal, QC H3A 0G1, Canada; (K.I.G.); (M.M.); (A.P.); (A.J.N.); (M.A.); (A.S.); (P.J.)
- Department of Human Genetics, McGill University, 1205 Doctor Penfield Avenue, Montreal, QC H3A 1B1, Canada
- Correspondence:
| |
Collapse
|
5
|
Reinfeld BI, Madden MZ, Wolf MM, Chytil A, Bader JE, Patterson AR, Sugiura A, Cohen AS, Ali A, Do BT, Muir A, Lewis CA, Hongo RA, Young KL, Brown RE, Todd VM, Huffstater T, Abraham A, O'Neil RT, Wilson MH, Xin F, Tantawy MN, Merryman WD, Johnson RW, Williams CS, Mason EF, Mason FM, Beckermann KE, Vander Heiden MG, Manning HC, Rathmell JC, Rathmell WK. Cell-programmed nutrient partitioning in the tumour microenvironment. Nature 2021; 593:282-288. [PMID: 33828302 PMCID: PMC8122068 DOI: 10.1038/s41586-021-03442-1] [Citation(s) in RCA: 543] [Impact Index Per Article: 181.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Accepted: 03/10/2021] [Indexed: 02/01/2023]
Abstract
Cancer cells characteristically consume glucose through Warburg metabolism1, a process that forms the basis of tumour imaging by positron emission tomography (PET). Tumour-infiltrating immune cells also rely on glucose, and impaired immune cell metabolism in the tumour microenvironment (TME) contributes to immune evasion by tumour cells2-4. However, whether the metabolism of immune cells is dysregulated in the TME by cell-intrinsic programs or by competition with cancer cells for limited nutrients remains unclear. Here we used PET tracers to measure the access to and uptake of glucose and glutamine by specific cell subsets in the TME. Notably, myeloid cells had the greatest capacity to take up intratumoral glucose, followed by T cells and cancer cells, across a range of cancer models. By contrast, cancer cells showed the highest uptake of glutamine. This distinct nutrient partitioning was programmed in a cell-intrinsic manner through mTORC1 signalling and the expression of genes related to the metabolism of glucose and glutamine. Inhibiting glutamine uptake enhanced glucose uptake across tumour-resident cell types, showing that glutamine metabolism suppresses glucose uptake without glucose being a limiting factor in the TME. Thus, cell-intrinsic programs drive the preferential acquisition of glucose and glutamine by immune and cancer cells, respectively. Cell-selective partitioning of these nutrients could be exploited to develop therapies and imaging strategies to enhance or monitor the metabolic programs and activities of specific cell populations in the TME.
Collapse
Affiliation(s)
- Bradley I Reinfeld
- Medical Scientist Training Program, Vanderbilt University, Nashville, TN, USA
- Department of Medicine, Vanderbilt University Medical Center (VUMC), Nashville, TN, USA
- Graduate Program in Cancer Biology, Vanderbilt University, Nashville, TN, USA
| | - Matthew Z Madden
- Medical Scientist Training Program, Vanderbilt University, Nashville, TN, USA
- Department of Pathology, Microbiology and Immunology, VUMC, Nashville, TN, USA
| | - Melissa M Wolf
- Department of Medicine, Vanderbilt University Medical Center (VUMC), Nashville, TN, USA
- Graduate Program in Cancer Biology, Vanderbilt University, Nashville, TN, USA
| | - Anna Chytil
- Department of Medicine, Vanderbilt University Medical Center (VUMC), Nashville, TN, USA
| | - Jackie E Bader
- Department of Pathology, Microbiology and Immunology, VUMC, Nashville, TN, USA
| | - Andrew R Patterson
- Department of Pathology, Microbiology and Immunology, VUMC, Nashville, TN, USA
| | - Ayaka Sugiura
- Medical Scientist Training Program, Vanderbilt University, Nashville, TN, USA
- Department of Pathology, Microbiology and Immunology, VUMC, Nashville, TN, USA
| | - Allison S Cohen
- Department of Radiology and Radiological Sciences, VUMC, Nashville, TN, USA
- Vanderbilt University Institute of Imaging Science, VUMC, Nashville, TN, USA
| | - Ahmed Ali
- Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Brian T Do
- Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Alexander Muir
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA
| | - Caroline A Lewis
- Whitehead Institute for Biomedical Research, MIT, Cambridge, MA, USA
| | - Rachel A Hongo
- Department of Medicine, Vanderbilt University Medical Center (VUMC), Nashville, TN, USA
- Department of Pathology, Microbiology and Immunology, VUMC, Nashville, TN, USA
| | - Kirsten L Young
- Department of Medicine, Vanderbilt University Medical Center (VUMC), Nashville, TN, USA
- Department of Pathology, Microbiology and Immunology, VUMC, Nashville, TN, USA
| | - Rachel E Brown
- Medical Scientist Training Program, Vanderbilt University, Nashville, TN, USA
- Department of Medicine, Vanderbilt University Medical Center (VUMC), Nashville, TN, USA
- Graduate Program in Cancer Biology, Vanderbilt University, Nashville, TN, USA
| | - Vera M Todd
- Department of Medicine, Vanderbilt University Medical Center (VUMC), Nashville, TN, USA
- Graduate Program in Cancer Biology, Vanderbilt University, Nashville, TN, USA
| | - Tessa Huffstater
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Abin Abraham
- Medical Scientist Training Program, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Genetics Institute, VUMC, Nashville, TN, USA
| | - Richard T O'Neil
- Department of Medicine, Vanderbilt University Medical Center (VUMC), Nashville, TN, USA
- Department of Veterans Affairs, Tennessee Valley Health System, Nashville, TN, USA
| | - Matthew H Wilson
- Department of Medicine, Vanderbilt University Medical Center (VUMC), Nashville, TN, USA
- Department of Veterans Affairs, Tennessee Valley Health System, Nashville, TN, USA
| | - Fuxue Xin
- Department of Radiology and Radiological Sciences, VUMC, Nashville, TN, USA
- Vanderbilt University Institute of Imaging Science, VUMC, Nashville, TN, USA
| | - M Noor Tantawy
- Department of Radiology and Radiological Sciences, VUMC, Nashville, TN, USA
- Vanderbilt University Institute of Imaging Science, VUMC, Nashville, TN, USA
| | - W David Merryman
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Rachelle W Johnson
- Department of Medicine, Vanderbilt University Medical Center (VUMC), Nashville, TN, USA
| | - Christopher S Williams
- Department of Medicine, Vanderbilt University Medical Center (VUMC), Nashville, TN, USA
- Department of Veterans Affairs, Tennessee Valley Health System, Nashville, TN, USA
| | - Emily F Mason
- Department of Pathology, Microbiology and Immunology, VUMC, Nashville, TN, USA
| | - Frank M Mason
- Department of Medicine, Vanderbilt University Medical Center (VUMC), Nashville, TN, USA
| | | | - Matthew G Vander Heiden
- Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
- Broad Institute of MIT and Harvard University, Cambridge, MA, USA
- Dana-Farber Cancer Institute, Boston, MA, USA
| | - H Charles Manning
- Department of Radiology and Radiological Sciences, VUMC, Nashville, TN, USA
- Vanderbilt University Institute of Imaging Science, VUMC, Nashville, TN, USA
| | - Jeffrey C Rathmell
- Department of Pathology, Microbiology and Immunology, VUMC, Nashville, TN, USA.
- Vanderbilt Center for Immunobiology and Vanderbilt-Ingram Cancer Center, VUMC, Nashville, TN, USA.
| | - W Kimryn Rathmell
- Department of Medicine, Vanderbilt University Medical Center (VUMC), Nashville, TN, USA.
- Vanderbilt Center for Immunobiology and Vanderbilt-Ingram Cancer Center, VUMC, Nashville, TN, USA.
| |
Collapse
|
6
|
Trott JF, Abu Aboud O, McLaughlin B, Anderson KL, Modiano JF, Kim K, Jen KY, Senapedis W, Chang H, Landesman Y, Baloglu E, Pili R, Weiss RH. Anti-Cancer Activity of PAK4/NAMPT Inhibitor and Programmed Cell Death Protein-1 Antibody in Kidney Cancer. KIDNEY360 2020; 1:376-388. [PMID: 35224510 PMCID: PMC8809296 DOI: 10.34067/kid.0000282019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 03/12/2020] [Indexed: 06/14/2023]
Abstract
BACKGROUND Kidney cancer (or renal cell carcinoma, RCC) is the sixth most common malignancy in the United States and is increasing in incidence. Despite new therapies, including targeted therapies and immunotherapies, most RCCs are resistant to treatment. Thus, several laboratories have been evaluating new approaches to therapy, both with single agents as well as combinations. Although we have previously shown efficacy of the dual PAK4/nicotinamide phosphoribosyltransferase (NAMPT) inhibitor KPT-9274, and the immune checkpoint inhibitors (CPI) have shown utility in the clinic, there has been no evaluation of this combination either clinically or in an immunocompetent animal model of kidney cancer. METHODS In this study, we use the renal cell adenocarcinoma (RENCA) model of spontaneous murine kidney cancer. Male BALB/cJ mice were injected subcutaneously with RENCA cells and, after tumors were palpable, they were treated with KPT-9274 and/or anti-programmed cell death 1 (PDCD1; PD1) antibody for 21 days. Tumors were measured and then removed at animal euthanasia for subsequent studies. RESULTS We demonstrate a significant decrease in allograft growth with the combination treatment of KPT-9274 and anti-PD1 antibody without significant weight loss by the animals. This is associated with decreased (MOUSE) Naprt expression, indicating dependence of these tumors on NAMPT in parallel to what we have observed in human RCC. Histology of the tumors showed substantial necrosis regardless of treatment condition, and flow cytometry of antibody-stained tumor cells revealed that the enhanced therapeutic effect of KPT-9274 and anti-PD1 antibody was not driven by infiltration of T cells into tumors. CONCLUSIONS This study highlights the potential of the RENCA model for evaluating immunologic responses to KPT-9274 and checkpoint inhibitor (CPI) and suggests that therapy with this combination could improve efficacy in RCC beyond what is achievable with CPI alone.
Collapse
Affiliation(s)
- Josephine F. Trott
- Division of Nephrology, Department of Internal Medicine, University of California, Davis, California
| | - Omran Abu Aboud
- Division of Nephrology, Department of Internal Medicine, University of California, Davis, California
| | - Bridget McLaughlin
- Comprehensive Cancer Center, University of California, Davis, California
| | - Katie L. Anderson
- Animal Cancer Care and Research Program, College of Veterinary Medicine, University of Minnesota, St Paul, Minnesota
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St Paul, Minnesota
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
- Center for Immunology, University of Minnesota, Minneapolis, Minnesota
| | - Jaime F. Modiano
- Animal Cancer Care and Research Program, College of Veterinary Medicine, University of Minnesota, St Paul, Minnesota
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St Paul, Minnesota
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
- Center for Immunology, University of Minnesota, Minneapolis, Minnesota
- Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota
| | - Kyoungmi Kim
- Division of Biostatistics, Department of Public Health Sciences, University of California, Davis, California
| | - Kuang-Yu Jen
- Department of Pathology and Laboratory Medicine, University of California, Davis, California
| | - William Senapedis
- Research and Translational Development, Karyopharm Therapeutics Inc., Newton, Massachusetts
| | - Hua Chang
- Research and Translational Development, Karyopharm Therapeutics Inc., Newton, Massachusetts
| | - Yosef Landesman
- Research and Translational Development, Karyopharm Therapeutics Inc., Newton, Massachusetts
| | - Erkan Baloglu
- Research and Translational Development, Karyopharm Therapeutics Inc., Newton, Massachusetts
| | - Roberto Pili
- Simon Cancer Center, School of Medicine, Indiana University, Indianapolis, Indiana
| | - Robert H. Weiss
- Division of Nephrology, Department of Internal Medicine, University of California, Davis, California
- Comprehensive Cancer Center, University of California, Davis, California
- Medical Service, Veterans Affairs Northern California Health Care System, Sacramento, California
| |
Collapse
|
7
|
Sobczuk P, Brodziak A, Khan MI, Chhabra S, Fiedorowicz M, Wełniak-Kamińska M, Synoradzki K, Bartnik E, Cudnoch-Jędrzejewska A, Czarnecka AM. Choosing The Right Animal Model for Renal Cancer Research. Transl Oncol 2020; 13:100745. [PMID: 32092671 PMCID: PMC7036425 DOI: 10.1016/j.tranon.2020.100745] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 01/04/2020] [Accepted: 01/06/2020] [Indexed: 12/17/2022] Open
Abstract
The increase in the life expectancy of patients with renal cell carcinoma (RCC) in the last decade is due to changes that have occurred in the area of preclinical studies. Understanding cancer pathophysiology and the emergence of new therapeutic options, including immunotherapy, would not be possible without proper research. Before new approaches to disease treatment are developed and introduced into clinical practice they must be preceded by preclinical tests, in which animal studies play a significant role. This review describes the progress in animal model development in kidney cancer research starting from the oldest syngeneic or chemically-induced models, through genetically modified mice, finally to xenograft, especially patient-derived, avatar and humanized mouse models. As there are a number of subtypes of RCC, our aim is to help to choose the right animal model for a particular kidney cancer subtype. The data on genetic backgrounds, biochemical parameters, histology, different stages of carcinogenesis and metastasis in various animal models of RCC as well as their translational relevance are summarized. Moreover, we shed some light on imaging methods, which can help define tumor microstructure, assist in the analysis of its metabolic changes and track metastasis development.
Collapse
Affiliation(s)
- Paweł Sobczuk
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland; Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Warsaw, Poland.
| | - Anna Brodziak
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland; Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Warsaw, Poland.
| | - Mohammed Imran Khan
- Department of Otolaryngology - Head & Neck Surgery, Western University, London, Ontario, Canada.
| | - Stuti Chhabra
- Department of Biochemistry, CSIR-Central Drug Research Institute, Lucknow, India.
| | - Michał Fiedorowicz
- Department of Experimental Pharmacology, Mossakowski Medical Research Centre Polish Academy of Sciences, 5 Pawinskiego Str., Warsaw, Poland.
| | - Marlena Wełniak-Kamińska
- Department of Experimental Pharmacology, Mossakowski Medical Research Centre Polish Academy of Sciences, 5 Pawinskiego Str., Warsaw, Poland.
| | - Kamil Synoradzki
- Department of Experimental Pharmacology, Mossakowski Medical Research Centre Polish Academy of Sciences, 5 Pawinskiego Str., Warsaw, Poland.
| | - Ewa Bartnik
- Institute of Genetics and Biotechnology, Faculty of Biology, University of Warsaw, Warsaw, Poland.
| | - Agnieszka Cudnoch-Jędrzejewska
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland.
| | - Anna M Czarnecka
- Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Warsaw, Poland; Department of Experimental Pharmacology, Mossakowski Medical Research Centre Polish Academy of Sciences, 5 Pawinskiego Str., Warsaw, Poland.
| |
Collapse
|
8
|
Enhanced efficacy of sitravatinib in metastatic models of antiangiogenic therapy resistance. PLoS One 2019; 14:e0220101. [PMID: 31369645 PMCID: PMC6675057 DOI: 10.1371/journal.pone.0220101] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 07/08/2019] [Indexed: 01/09/2023] Open
Abstract
Tyrosine kinase inhibitors (TKIs) that primarily target angiogenesis are approved to treat several cancers in the metastatic setting; however, resistance is common. Sequential treatment or 'switching' from one TKI to another following failure can be effective, but predicting which drugs will have cross-over sensitivity remains a challenge. Here we examined sitravatinib (MGCD516), a spectrum-selective TKI able to block MET, TAM (TYRO3, AXL, MerTK) and multiple receptor families (including PDGFRs, VEGFRs, and Ephs). Transcriptomic analysis of several mouse and human cell lines revealed diverse molecular changes after resistance to two TKIs (sunitinib and axitinib) with multiple sitravatinib targets found to be upregulated. Sitravatinib treatment in vitro resulted in enhanced anti-proliferative effects in resistant cells and was improved compared to TKIs with similar target profiles. In vivo, primary tumor growth inhibition after sitravatinib treatment in mice was enhanced in resistant tumors and metastasis suppression improved when tumors were surgically removed. Together, these results suggest that the diverse and often inconsistent compensatory signaling mechanisms found to contribute to TKI resistance may paradoxically improve the tumor-inhibiting effects of broad-spectrum TKIs such as sitravatinib that are able to block multiple signaling pathways. Sitravatinib in the second-line setting following antiangiogenic TKI treatment may have enhanced inhibitory effects in local and disseminated disease, and improve outcomes in patients with refractory disease.
Collapse
|
9
|
Yeku O, Li X, Brentjens RJ. Adoptive T-Cell Therapy for Solid Tumors. AMERICAN SOCIETY OF CLINICAL ONCOLOGY EDUCATIONAL BOOK. AMERICAN SOCIETY OF CLINICAL ONCOLOGY. ANNUAL MEETING 2017. [PMID: 28561728 DOI: 10.14694/edbk_180328] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy is an innovative form of immunotherapy wherein autologous T cells are genetically modified to express chimeric receptors encoding an antigen-specific single-chain variable fragment and various costimulatory molecules. Upon administration, these modified T cells traffic to, and recognize, cancer cells in an HLA-independent manner. CAR T-cell therapy has shown remarkable success in the treatment of CD-19-expressing B-cell acute lymphocytic leukemia. However, clinical gains to the same magnitude have not been reported in solid tumors. Several known obstacles to CAR T-cell therapy for solid tumors include target antigen identification, effective trafficking to the tumor, robust activation, proliferation, and in vivo cytotoxicity. Beyond these T-cell intrinsic properties, a complex and dynamic immunosuppressive tumor microenvironment in solid tumors hinders T-cell efficacy. Notable advancements in CAR design to include multiple costimulatory molecules, ligands, and soluble cytokines have shown promise in preclinical models, and some of these are currently in early-phase clinical trials. In this review, we discuss selected solid tumor malignancies and relevant preclinical data and highlight clinical trial results that are available. Furthermore, we outline some obstacles to CAR T-cell therapy for each tumor and propose strategies to overcome some of these limitations.
Collapse
Affiliation(s)
- Oladapo Yeku
- From the Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY; Weill Cornell Medicine, New York, NY; Center for Cell Engineering, and Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Xinghuo Li
- From the Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY; Weill Cornell Medicine, New York, NY; Center for Cell Engineering, and Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Renier J Brentjens
- From the Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY; Weill Cornell Medicine, New York, NY; Center for Cell Engineering, and Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
10
|
Yeku O, Li X, Brentjens RJ. Adoptive T-Cell Therapy for Solid Tumors. Am Soc Clin Oncol Educ Book 2017; 37:193-204. [PMID: 28561728 DOI: 10.1200/edbk_180328] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy is an innovative form of immunotherapy wherein autologous T cells are genetically modified to express chimeric receptors encoding an antigen-specific single-chain variable fragment and various costimulatory molecules. Upon administration, these modified T cells traffic to, and recognize, cancer cells in an HLA-independent manner. CAR T-cell therapy has shown remarkable success in the treatment of CD-19-expressing B-cell acute lymphocytic leukemia. However, clinical gains to the same magnitude have not been reported in solid tumors. Several known obstacles to CAR T-cell therapy for solid tumors include target antigen identification, effective trafficking to the tumor, robust activation, proliferation, and in vivo cytotoxicity. Beyond these T-cell intrinsic properties, a complex and dynamic immunosuppressive tumor microenvironment in solid tumors hinders T-cell efficacy. Notable advancements in CAR design to include multiple costimulatory molecules, ligands, and soluble cytokines have shown promise in preclinical models, and some of these are currently in early-phase clinical trials. In this review, we discuss selected solid tumor malignancies and relevant preclinical data and highlight clinical trial results that are available. Furthermore, we outline some obstacles to CAR T-cell therapy for each tumor and propose strategies to overcome some of these limitations.
Collapse
Affiliation(s)
- Oladapo Yeku
- From the Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY; Weill Cornell Medicine, New York, NY; Center for Cell Engineering, and Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Xinghuo Li
- From the Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY; Weill Cornell Medicine, New York, NY; Center for Cell Engineering, and Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Renier J Brentjens
- From the Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY; Weill Cornell Medicine, New York, NY; Center for Cell Engineering, and Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
11
|
Falls T, Roy DG, Bell JC, Bourgeois-Daigneault MC. Murine Tumor Models for Oncolytic Rhabdo-Virotherapy. ILAR J 2017; 57:73-85. [PMID: 27034397 DOI: 10.1093/ilar/ilv048] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The preclinical optimization and validation of novel treatments for cancer therapy requires the use of laboratory animals. Although in vitro experiments using tumor cell lines and ex vivo treatment of patient tumor samples provide a remarkable first-line tool for the initial study of tumoricidal potential, tumor-bearing animals remain the primary option to study delivery, efficacy, and safety of therapies in the context of a complete tumor microenvironment and functional immune system. In this review, we will describe the use of murine tumor models for oncolytic virotherapy using vesicular stomatitis virus. We will discuss studies using immunocompetent and immunodeficient models with respect to toxicity and therapeutic treatments, as well as the various techniques and tools available to study cancer therapy with Rhabdoviruses.
Collapse
Affiliation(s)
- Theresa Falls
- Theresa Falls is a research technician at the Centre for Innovative Cancer Research at Ottawa Hospital Research Institute in Ottawa, Canada. Dominic Guy Roy is a Ph.D candidate at the Centre for Innovative Cancer Research at Ottawa Hospital Research Institute in Ottawa, Canada, and a Ph.D candidate in the Biochemistry, Microbiology, and Immunology Department at the University of Ottawa in Ottawa, Canada. John Cameron Bell is a senior researcher at the Centre for Innovative Cancer Research at Ottawa Hospital Research Institute in Ottawa, Canada, and professor in the Biochemistry, Microbiology, and Immunology Department at the University of Ottawa in Ottawa, Canada. Marie-Claude Bourgeois-Daigneault is a postdoctoral fellow at the Centre for Innovative Cancer Research at Ottawa Hospital Research Institute in Ottawa, Canada, and a postdoctoral fellow in the Biochemistry, Microbiology, and Immunology Department at the University of Ottawa in Ottawa, Canada
| | - Dominic Guy Roy
- Theresa Falls is a research technician at the Centre for Innovative Cancer Research at Ottawa Hospital Research Institute in Ottawa, Canada. Dominic Guy Roy is a Ph.D candidate at the Centre for Innovative Cancer Research at Ottawa Hospital Research Institute in Ottawa, Canada, and a Ph.D candidate in the Biochemistry, Microbiology, and Immunology Department at the University of Ottawa in Ottawa, Canada. John Cameron Bell is a senior researcher at the Centre for Innovative Cancer Research at Ottawa Hospital Research Institute in Ottawa, Canada, and professor in the Biochemistry, Microbiology, and Immunology Department at the University of Ottawa in Ottawa, Canada. Marie-Claude Bourgeois-Daigneault is a postdoctoral fellow at the Centre for Innovative Cancer Research at Ottawa Hospital Research Institute in Ottawa, Canada, and a postdoctoral fellow in the Biochemistry, Microbiology, and Immunology Department at the University of Ottawa in Ottawa, Canada
| | - John Cameron Bell
- Theresa Falls is a research technician at the Centre for Innovative Cancer Research at Ottawa Hospital Research Institute in Ottawa, Canada. Dominic Guy Roy is a Ph.D candidate at the Centre for Innovative Cancer Research at Ottawa Hospital Research Institute in Ottawa, Canada, and a Ph.D candidate in the Biochemistry, Microbiology, and Immunology Department at the University of Ottawa in Ottawa, Canada. John Cameron Bell is a senior researcher at the Centre for Innovative Cancer Research at Ottawa Hospital Research Institute in Ottawa, Canada, and professor in the Biochemistry, Microbiology, and Immunology Department at the University of Ottawa in Ottawa, Canada. Marie-Claude Bourgeois-Daigneault is a postdoctoral fellow at the Centre for Innovative Cancer Research at Ottawa Hospital Research Institute in Ottawa, Canada, and a postdoctoral fellow in the Biochemistry, Microbiology, and Immunology Department at the University of Ottawa in Ottawa, Canada
| | - Marie-Claude Bourgeois-Daigneault
- Theresa Falls is a research technician at the Centre for Innovative Cancer Research at Ottawa Hospital Research Institute in Ottawa, Canada. Dominic Guy Roy is a Ph.D candidate at the Centre for Innovative Cancer Research at Ottawa Hospital Research Institute in Ottawa, Canada, and a Ph.D candidate in the Biochemistry, Microbiology, and Immunology Department at the University of Ottawa in Ottawa, Canada. John Cameron Bell is a senior researcher at the Centre for Innovative Cancer Research at Ottawa Hospital Research Institute in Ottawa, Canada, and professor in the Biochemistry, Microbiology, and Immunology Department at the University of Ottawa in Ottawa, Canada. Marie-Claude Bourgeois-Daigneault is a postdoctoral fellow at the Centre for Innovative Cancer Research at Ottawa Hospital Research Institute in Ottawa, Canada, and a postdoctoral fellow in the Biochemistry, Microbiology, and Immunology Department at the University of Ottawa in Ottawa, Canada
| |
Collapse
|
12
|
Schokrpur S, Hu J, Moughon DL, Liu P, Lin LC, Hermann K, Mangul S, Guan W, Pellegrini M, Xu H, Wu L. CRISPR-Mediated VHL Knockout Generates an Improved Model for Metastatic Renal Cell Carcinoma. Sci Rep 2016; 6:29032. [PMID: 27358011 PMCID: PMC4928183 DOI: 10.1038/srep29032] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 06/14/2016] [Indexed: 12/15/2022] Open
Abstract
Metastatic renal cell carcinoma (mRCC) is nearly incurable and accounts for most of the mortality associated with RCC. Von Hippel Lindau (VHL) is a tumour suppressor that is lost in the majority of clear cell RCC (ccRCC) cases. Its role in regulating hypoxia-inducible factors-1α (HIF-1α) and -2α (HIF-2α) is well-studied. Recent work has demonstrated that VHL knock down induces an epithelial-mesenchymal transition (EMT) phenotype. In this study we showed that a CRISPR/Cas9-mediated knock out of VHL in the RENCA model leads to morphologic and molecular changes indicative of EMT, which in turn drives increased metastasis to the lungs. RENCA cells deficient in HIF-1α failed to undergo EMT changes upon VHL knockout. RNA-seq revealed several HIF-1α-regulated genes that are upregulated in our VHL knockout cells and whose overexpression signifies an aggressive form of ccRCC in the cancer genome atlas (TCGA) database. Independent validation in a new clinical dataset confirms the upregulation of these genes in ccRCC samples compared to adjacent normal tissue. Our findings indicate that loss of VHL could be driving tumour cell dissemination through stabilization of HIF-1α in RCC. A better understanding of the mechanisms involved in this phenomenon can guide the search for more effective treatments to combat mRCC.
Collapse
MESH Headings
- Animals
- Bacterial Proteins
- CRISPR-Associated Protein 9
- CRISPR-Cas Systems
- Carcinoma, Renal Cell/genetics
- Carcinoma, Renal Cell/secondary
- Cell Line, Tumor
- Cell Movement
- Datasets as Topic
- Disease Models, Animal
- Endonucleases
- Epithelial-Mesenchymal Transition
- Female
- Gene Editing
- Gene Expression Regulation, Neoplastic
- Heterografts
- Humans
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- Hypoxia-Inducible Factor 1, alpha Subunit/physiology
- Kidney Neoplasms/pathology
- Lung Neoplasms/secondary
- Mice
- Mice, Knockout
- Mice, Nude
- Neoplasm Proteins/genetics
- Neoplasm Proteins/physiology
- RNA, Guide, CRISPR-Cas Systems
- Von Hippel-Lindau Tumor Suppressor Protein/genetics
- Von Hippel-Lindau Tumor Suppressor Protein/physiology
Collapse
Affiliation(s)
- Shiruyeh Schokrpur
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California at Los Angeles CA 90095, USA
| | - Junhui Hu
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California at Los Angeles CA 90095, USA
- Department of Urology and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Department of Paediatric Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Diana L. Moughon
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California at Los Angeles CA 90095, USA
| | - Peijun Liu
- Department of Urology and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Lucia C. Lin
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California at Los Angeles CA 90095, USA
| | - Kip Hermann
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California at Los Angeles CA 90095, USA
| | - Serghei Mangul
- Department of Computer Science and Human Genetics, University of California at Los Angeles CA 90095, USA
| | - Wei Guan
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California at Los Angeles CA 90095, USA
- Department of Urology and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Matteo Pellegrini
- Department of Computer Science and Human Genetics, University of California at Los Angeles CA 90095, USA
- Department of Molecular, Cell, and Developmental Biology, University of California at Los Angeles CA 90095, USA
| | - Hua Xu
- Department of Urology and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Lily Wu
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California at Los Angeles CA 90095, USA
- Department of Urology, David Geffen School of Medicine, University of California at Los Angeles CA 90095, USA
| |
Collapse
|
13
|
Benzekry S, Tracz A, Mastri M, Corbelli R, Barbolosi D, Ebos JML. Modeling Spontaneous Metastasis following Surgery: An In Vivo-In Silico Approach. Cancer Res 2015; 76:535-47. [PMID: 26511632 DOI: 10.1158/0008-5472.can-15-1389] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Accepted: 09/29/2015] [Indexed: 12/19/2022]
Abstract
Rapid improvements in the detection and tracking of early-stage tumor progression aim to guide decisions regarding cancer treatments as well as predict metastatic recurrence in patients following surgery. Mathematical models may have the potential to further assist in estimating metastatic risk, particularly when paired with in vivo tumor data that faithfully represent all stages of disease progression. Herein, we describe mathematical analysis that uses data from mouse models of spontaneous metastasis developing after surgical removal of orthotopically implanted primary tumors. Both presurgical (primary tumor) growth and postsurgical (metastatic) growth were quantified using bioluminescence and were then used to generate a mathematical formalism based on general laws of the disease (i.e., dissemination and growth). The model was able to fit and predict pre/postsurgical data at the level of the individual as well as the population. Our approach also enabled retrospective analysis of clinical data describing the probability of metastatic relapse as a function of primary tumor size. In these data-based models, interindividual variability was quantified by a key parameter of intrinsic metastatic potential. Critically, our analysis identified a highly nonlinear relationship between primary tumor size and postsurgical survival, suggesting possible threshold limits for the utility of tumor size as a predictor of metastatic recurrence. These findings represent a novel use of clinically relevant models to assess the impact of surgery on metastatic potential and may guide optimal timing of treatments in neoadjuvant (presurgical) and adjuvant (postsurgical) settings to maximize patient benefit.
Collapse
Affiliation(s)
- Sebastien Benzekry
- Inria Bordeaux Sud-Ouest, Team MONC, Institut de Mathematiques de Bordeaux, Bordeaux, France.
| | - Amanda Tracz
- Department of Cancer Genetics, Roswell Park Cancer Institute, Buffalo, New York
| | - Michalis Mastri
- Department of Cancer Genetics, Roswell Park Cancer Institute, Buffalo, New York
| | - Ryan Corbelli
- Department of Cancer Genetics, Roswell Park Cancer Institute, Buffalo, New York
| | - Dominique Barbolosi
- SMARTc Pharmacokinetics Unit, Inserm S 911 CRO2, Aix Marseille University, Marseille, France
| | - John M L Ebos
- Department of Cancer Genetics, Roswell Park Cancer Institute, Buffalo, New York. Department of Medicine, Roswell Park Cancer Institute, Buffalo, New York
| |
Collapse
|
14
|
Ebos JML, Mastri M, Lee CR, Tracz A, Hudson JM, Attwood K, Cruz-Munoz WR, Jedeszko C, Burns P, Kerbel RS. Neoadjuvant antiangiogenic therapy reveals contrasts in primary and metastatic tumor efficacy. EMBO Mol Med 2015; 6:1561-76. [PMID: 25361689 PMCID: PMC4287975 DOI: 10.15252/emmm.201403989] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Thousands of cancer patients are currently in clinical trials evaluating antiangiogenic therapy
in the neoadjuvant setting, which is the treatment of localized primary tumors prior to surgical
intervention. The rationale is that shrinking a tumor will improve surgical outcomes and minimize
growth of occult micrometastatic disease—thus delaying post-surgical recurrence and improving
survival. But approved VEGF pathway inhibitors have not been tested in clinically relevant
neoadjuvant models that compare pre- and post-surgical treatment effects. Using mouse models of
breast, kidney, and melanoma metastasis, we demonstrate that primary tumor responses to neoadjuvant
VEGFR TKI treatment do not consistently correlate with improved post-surgical survival, with
survival worsened in certain settings. Similar negative effects did not extend to protein-based VEGF
pathway inhibitors and could be reversed with altered dose, surgical timing, and treatment duration,
or when VEGFR TKIs are combined with metronomic ‘anti-metastatic’ chemotherapy
regimens. These studies represent the first attempt to recapitulate the complex clinical parameters
of neoadjuvant therapy in mice and identify a novel tool to compare systemic antiangiogenic
treatment effects on localized and disseminated disease.
Collapse
Affiliation(s)
- John M L Ebos
- Genitourinary Section, Department of Medicine, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Michalis Mastri
- Genitourinary Section, Department of Medicine, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Christina R Lee
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Amanda Tracz
- Genitourinary Section, Department of Medicine, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - John M Hudson
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Kristopher Attwood
- Department of Biostatistics and Bioinformatics, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - William R Cruz-Munoz
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Christopher Jedeszko
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Peter Burns
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, ON, Canada Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Robert S Kerbel
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, ON, Canada Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
15
|
Lodhia KA, Hadley AM, Haluska P, Scott CL. Prioritizing therapeutic targets using patient-derived xenograft models. Biochim Biophys Acta Rev Cancer 2015; 1855:223-34. [PMID: 25783201 DOI: 10.1016/j.bbcan.2015.03.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Revised: 02/12/2015] [Accepted: 03/09/2015] [Indexed: 01/03/2023]
Abstract
Effective systemic treatment of cancer relies on the delivery of agents with optimal therapeutic potential. The molecular age of medicine has provided genomic tools that can identify a large number of potential therapeutic targets in individual patients, heralding the promise of personalized treatment. However, determining which potential targets actually drive tumor growth and should be prioritized for therapy is challenging. Indeed, reliable molecular matches of target and therapeutic agent have been stringently validated in the clinic for only a small number of targets. Patient-derived xenografts (PDXs) are tumor models developed in immunocompromised mice using tumor procured directly from the patient. As patient surrogates, PDX models represent a powerful tool for addressing individualized therapy. Challenges include humanizing the immune system of PDX models and ensuring high quality molecular annotation, in order to maximize insights for the clinic. Importantly, PDX can be sampled repeatedly and in parallel, to reveal clonal evolution, which may predict mechanisms of drug resistance and inform therapeutic strategy design.
Collapse
Affiliation(s)
- K A Lodhia
- Department of Oncology, Mayo Clinic, Rochester, MN, USA
| | - A M Hadley
- Stem Cells and Cancer Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
| | - P Haluska
- Department of Oncology, Mayo Clinic, Rochester, MN, USA
| | - C L Scott
- Stem Cells and Cancer Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|