1
|
Stransky LA, Gao W, Schmidt LS, Bi K, Ricketts CJ, Ramesh V, James A, Difilippantonio S, Ileva L, Kalen JD, Karim B, Jeon A, Morgan T, Warner AC, Turan S, Unite J, Tran B, Choudhari S, Zhao Y, Linn DE, Yun C, Dhandapani S, Parab V, Pinheiro EM, Morris N, He L, Vigeant SM, Pignon JC, Sticco-Ivins M, Signoretti S, Van Allen EM, Linehan WM, Kaelin WG. Toward a CRISPR-based mouse model of Vhl-deficient clear cell kidney cancer: Initial experience and lessons learned. Proc Natl Acad Sci U S A 2024; 121:e2408549121. [PMID: 39365820 PMCID: PMC11474080 DOI: 10.1073/pnas.2408549121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 09/03/2024] [Indexed: 10/06/2024] Open
Abstract
CRISPR is revolutionizing the ability to do somatic gene editing in mice for the purpose of creating new cancer models. Inactivation of the VHL tumor suppressor gene is the signature initiating event in the most common form of kidney cancer, clear cell renal cell carcinoma (ccRCC). Such tumors are usually driven by the excessive HIF2 activity that arises when the VHL gene product, pVHL, is defective. Given the pressing need for a robust immunocompetent mouse model of human ccRCC, we directly injected adenovirus-associated viruses (AAVs) encoding sgRNAs against VHL and other known/suspected ccRCC tumor suppressor genes into the kidneys of C57BL/6 mice under conditions where Cas9 was under the control of one of two different kidney-specific promoters (Cdh16 or Pax8) to induce kidney tumors. An AAV targeting Vhl, Pbrm1, Keap1, and Tsc1 reproducibly caused macroscopic ccRCCs that partially resembled human ccRCC tumors with respect to transcriptome and cell of origin and responded to a ccRCC standard-of-care agent, axitinib. Unfortunately, these tumors, like those produced by earlier genetically engineered mouse ccRCCs, are HIF2 independent.
Collapse
Affiliation(s)
- Laura A. Stransky
- Division of Molecular and Cellular Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA02215
| | - Wenhua Gao
- Division of Molecular and Cellular Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA02215
| | - Laura S. Schmidt
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD20892
- Basic Science Program, Frederick National Laboratory for Cancer Research, Frederick, MD21702
| | - Kevin Bi
- Division of Population Sciences, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA02115
- Center for Cancer Genomics, Dana-Farber Cancer Institute, Boston, MA02115
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA02142
| | - Christopher J. Ricketts
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD20892
| | - Vijyendra Ramesh
- Division of Molecular and Cellular Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA02215
| | - Amy James
- Animal Research Technical Support, Frederick National Laboratory for Cancer Research, Frederick, MD21702
| | - Simone Difilippantonio
- Animal Research Technical Support, Frederick National Laboratory for Cancer Research, Frederick, MD21702
| | - Lilia Ileva
- Small Animal Imaging Program, Frederick National Laboratory for Cancer Research, Frederick, MD21702
| | - Joseph D. Kalen
- Small Animal Imaging Program, Frederick National Laboratory for Cancer Research, Frederick, MD21702
| | - Baktiar Karim
- Molecular Histopathology Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD21702
| | - Albert Jeon
- Molecular Histopathology Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD21702
| | - Tamara Morgan
- Molecular Histopathology Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD21702
| | - Andrew C. Warner
- Molecular Histopathology Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD21702
| | - Sevilay Turan
- National Cancer Institute Center for Cancer Research, Sequencing Facility, Frederick National Laboratory for Cancer Research, Frederick, MD 21701
| | - Joanne Unite
- National Cancer Institute Center for Cancer Research, Sequencing Facility, Frederick National Laboratory for Cancer Research, Frederick, MD 21701
| | - Bao Tran
- National Cancer Institute Center for Cancer Research, Sequencing Facility, Frederick National Laboratory for Cancer Research, Frederick, MD 21701
| | - Sulbha Choudhari
- Advanced Biomedical and Computational Science, Frederick National Laboratory for Cancer Research, Frederick, MD21701
| | - Yongmei Zhao
- Advanced Biomedical and Computational Science, Frederick National Laboratory for Cancer Research, Frederick, MD21701
| | | | - Changhong Yun
- Pharmacokinetics, Merck & Co., Inc., Boston, MA02115
| | | | - Vaishali Parab
- Pharmacokinetics, Merck & Co., Inc., South San Francisco, CA94080
| | | | - Nicole Morris
- Laboratory of Animal Sciences Program, Frederick National Laboratory for Cancer Research, Frederick, MD21702
| | - Lixia He
- Division of Molecular and Cellular Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA02215
| | - Sean M. Vigeant
- Division of Molecular and Cellular Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA02215
| | - Jean-Christophe Pignon
- Harvard Medical School, Boston, MA02115
- Department of Pathology, Brigham and Women's Hospital, Boston, MA02115
| | - Maura Sticco-Ivins
- Harvard Medical School, Boston, MA02115
- Department of Pathology, Brigham and Women's Hospital, Boston, MA02115
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA02115
| | - Sabina Signoretti
- Harvard Medical School, Boston, MA02115
- Department of Pathology, Brigham and Women's Hospital, Boston, MA02115
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA02115
| | - Eliezer M. Van Allen
- Division of Population Sciences, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA02115
- Center for Cancer Genomics, Dana-Farber Cancer Institute, Boston, MA02115
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA02142
| | - W. Marston Linehan
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD20892
| | - William G. Kaelin
- Division of Molecular and Cellular Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA02215
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA02142
- HHMI, Chevy Chase, MD20815
| |
Collapse
|
2
|
Maksimovic S, Boscolo NC, La Posta L, Barrios S, Moussa MJ, Gentile E, Pesquera PI, Li W, Chen J, Gomez JA, Basi A, Burks JK, Alvarez-Breckenridge C, Gao J, Campbell MT, Dondossola E. Antiangiogenic Tyrosine Kinase Inhibitors have Differential Efficacy in Clear Cell Renal Cell Carcinoma in Bone. CANCER RESEARCH COMMUNICATIONS 2024; 4:2621-2637. [PMID: 39248577 PMCID: PMC11459607 DOI: 10.1158/2767-9764.crc-24-0304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 07/24/2024] [Accepted: 09/04/2024] [Indexed: 09/10/2024]
Abstract
Clear cell renal cell carcinoma (ccRCC) is the most prevalent kidney neoplasm; bone metastasis (BM) develops in 35% to 40% of metastatic patients and results in substantial morbidity and mortality, as well as medical costs. A key feature of ccRCC is the loss of function of the von Hippel-Lindau protein, which enhances angiogenesis via vascular endothelial growth factor release. Consequently, antiangiogenic tyrosine kinase inhibitors (TKI) emerged as a treatment for ccRCC. However, limited data about their efficacy in BM is available, and no systematic comparisons have been performed. We developed mouse models of bone and lung ccRCC tumors and compared their anticancer efficacy, impact on mouse survival, and mechanisms of action, including effects on tumor cells and both immune and nonimmune (blood vessels and osteoclasts) bone stromal components. This approach elucidates the efficacy of TKIs in ccRCC bone tumors to support rational interrogation and development of therapies. SIGNIFICANCE TKIs showed different efficacy in synchronous bone and lung metastases and did not eradicate tumors as single agents but induced extensive reprogramming of the BM microenvironment. This resulted in a significant decrease in neoangiogenic blood vessels, bone remodeling, and immune cell infiltration (including CD8 T cells) with altered spatial distribution.
Collapse
Affiliation(s)
- Stefan Maksimovic
- Department of Genitourinary Medical Oncology, David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, Texas.
- Division of Cancer Medicine, Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Nina C. Boscolo
- Department of Genitourinary Medical Oncology, David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, Texas.
- Division of Cancer Medicine, Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Ludovica La Posta
- Department of Genitourinary Medical Oncology, David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, Texas.
- Division of Cancer Medicine, Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Sergio Barrios
- Department of Genitourinary Medical Oncology, David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, Texas.
- Division of Cancer Medicine, Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
- Department of Bioengineering, Rice University, Houston, Texas.
| | - Mohammad Jad Moussa
- Division of Cancer Medicine, Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Emanuela Gentile
- Department of Genitourinary Medical Oncology, David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, Texas.
- Division of Cancer Medicine, Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Pedro I. Pesquera
- Division of Surgery, Department of Urology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Wenjiao Li
- Department of Genitourinary Medical Oncology, David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, Texas.
- Division of Cancer Medicine, Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Jianfeng Chen
- Department of Genitourinary Medical Oncology, David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, Texas.
- Division of Cancer Medicine, Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Javier A. Gomez
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Akshay Basi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Jared K. Burks
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | | | - Jianjun Gao
- Department of Genitourinary Medical Oncology, David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, Texas.
- Division of Cancer Medicine, Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Matthew T. Campbell
- Department of Genitourinary Medical Oncology, David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, Texas.
- Division of Cancer Medicine, Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Eleonora Dondossola
- Department of Genitourinary Medical Oncology, David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, Texas.
- Division of Cancer Medicine, Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
3
|
Voss MH, Motzer RJ. Adjuvant Immunotherapy for Kidney Cancer - A New Strategy with New Challenges. N Engl J Med 2024; 390:1432-1433. [PMID: 38631007 DOI: 10.1056/nejme2402364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/19/2024]
Affiliation(s)
- Martin H Voss
- From Memorial Sloan Kettering Cancer Center, New York
| | | |
Collapse
|
4
|
Wolf MM, Madden MZ, Arner EN, Bader JE, Ye X, Vlach L, Tigue ML, Landis MD, Jonker PB, Hatem Z, Steiner KK, Gaines DK, Reinfeld BI, Hathaway ES, Xin F, Tantawy MN, Haake SM, Jonasch E, Muir A, Weiss VL, Beckermann KE, Rathmell WK, Rathmell JC. VHL loss reprograms the immune landscape to promote an inflammatory myeloid microenvironment in renal tumorigenesis. J Clin Invest 2024; 134:e173934. [PMID: 38618956 PMCID: PMC11014672 DOI: 10.1172/jci173934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 02/24/2024] [Indexed: 04/16/2024] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is characterized by dysregulated hypoxia signaling and a tumor microenvironment (TME) highly enriched in myeloid and lymphoid cells. Loss of the von Hippel Lindau (VHL) gene is a critical early event in ccRCC pathogenesis and promotes stabilization of HIF. Whether VHL loss in cancer cells affects immune cells in the TME remains unclear. Using Vhl WT and Vhl-KO in vivo murine kidney cancer Renca models, we found that Vhl-KO tumors were more infiltrated by immune cells. Tumor-associated macrophages (TAMs) from Vhl-deficient tumors demonstrated enhanced in vivo glucose consumption, phagocytosis, and inflammatory transcriptional signatures, whereas lymphocytes from Vhl-KO tumors showed reduced activation and a lower response to anti-programmed cell death 1 (anti-PD-1) therapy in vivo. The chemokine CX3CL1 was highly expressed in human ccRCC tumors and was associated with Vhl deficiency. Deletion of Cx3cl1 in cancer cells decreased myeloid cell infiltration associated with Vhl loss to provide a mechanism by which Vhl loss may have contributed to the altered immune landscape. Here, we identify cancer cell-specific genetic features that drove environmental reprogramming and shaped the tumor immune landscape, with therapeutic implications for the treatment of ccRCC.
Collapse
Affiliation(s)
- Melissa M. Wolf
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville (VUMC), Tennessee, USA
- Graduate Program in Cancer Biology and
| | - Matthew Z. Madden
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville (VUMC), Tennessee, USA
- Medical Scientist Training Program, Vanderbilt University, Nashville, Tennessee, USA
| | - Emily N. Arner
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville (VUMC), Tennessee, USA
| | - Jackie E. Bader
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville (VUMC), Tennessee, USA
| | - Xiang Ye
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville (VUMC), Tennessee, USA
| | - Logan Vlach
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville (VUMC), Tennessee, USA
- Graduate Program in Cancer Biology and
| | - Megan L. Tigue
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville (VUMC), Tennessee, USA
- Graduate Program in Cancer Biology and
- Medical Scientist Training Program, Vanderbilt University, Nashville, Tennessee, USA
| | | | - Patrick B. Jonker
- Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois, USA
| | - Zaid Hatem
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville (VUMC), Tennessee, USA
| | - KayLee K. Steiner
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville (VUMC), Tennessee, USA
- Graduate Program in Cancer Biology and
| | - Dakim K. Gaines
- Department of Radiation Oncology
- Vanderbilt-Ingram Cancer Center
| | - Bradley I. Reinfeld
- Graduate Program in Cancer Biology and
- Medical Scientist Training Program, Vanderbilt University, Nashville, Tennessee, USA
- Department of Medicine, VUMC, Nashville, Tennessee, USA
| | - Emma S. Hathaway
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville (VUMC), Tennessee, USA
- Graduate Program in Cancer Biology and
| | - Fuxue Xin
- Department of Radiology and Radiological Sciences, and
- Vanderbilt University Institute of Imaging Science, VUMC, Nashville, Tennessee, USA
| | - M. Noor Tantawy
- Department of Radiology and Radiological Sciences, and
- Vanderbilt University Institute of Imaging Science, VUMC, Nashville, Tennessee, USA
| | - Scott M. Haake
- Department of Medicine, VUMC, Nashville, Tennessee, USA
- Vanderbilt-Ingram Cancer Center
| | - Eric Jonasch
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Alexander Muir
- Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois, USA
| | - Vivian L. Weiss
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville (VUMC), Tennessee, USA
- Vanderbilt-Ingram Cancer Center
| | - Kathryn E. Beckermann
- Department of Medicine, VUMC, Nashville, Tennessee, USA
- Vanderbilt-Ingram Cancer Center
| | - W. Kimryn Rathmell
- Department of Medicine, VUMC, Nashville, Tennessee, USA
- Vanderbilt-Ingram Cancer Center
- Vanderbilt Center for Immunobiology, VUMC, Nashville, Tennessee, USA
| | - Jeffrey C. Rathmell
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville (VUMC), Tennessee, USA
- Vanderbilt-Ingram Cancer Center
- Vanderbilt Center for Immunobiology, VUMC, Nashville, Tennessee, USA
| |
Collapse
|
5
|
Liao S, Wu G, Xie Z, Lei X, Yang X, Huang S, Deng X, Wang Z, Tang G. pH regulators and their inhibitors in tumor microenvironment. Eur J Med Chem 2024; 267:116170. [PMID: 38308950 DOI: 10.1016/j.ejmech.2024.116170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 01/14/2024] [Accepted: 01/22/2024] [Indexed: 02/05/2024]
Abstract
As an important characteristic of tumor, acidic tumor microenvironment (TME) is closely related to immune escape, invasion, migration and drug resistance of tumor. The acidity of the TME mainly comes from the acidic products produced by the high level of tumor metabolism, such as lactic acid and carbon dioxide. pH regulators such as monocarboxylate transporters (MCTs), carbonic anhydrase IX (CA IX), and Na+/H+ exchange 1 (NHE1) expel protons directly or indirectly from the tumor to maintain the pH balance of tumor cells and create an acidic TME. We review the functions of several pH regulators involved in the construction of acidic TME, the structure and structure-activity relationship of pH regulator inhibitors, and provide strategies for the development of small-molecule antitumor inhibitors based on these targets.
Collapse
Affiliation(s)
- Senyi Liao
- Institute of Pharmacy and Pharmacology, Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Guang Wu
- The Second Affiliated Hospital, Department of Pharmacy, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Zhizhong Xie
- Institute of Pharmacy and Pharmacology, Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Xiaoyong Lei
- Institute of Pharmacy and Pharmacology, Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Xiaoyan Yang
- Institute of Pharmacy and Pharmacology, Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Sheng Huang
- Jiuzhitang Co., Ltd, Changsha, Hunan, 410007, China
| | - Xiangping Deng
- The First Affiliated Hospital, Department of Pharmacy, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
| | - Zhe Wang
- The Second Affiliated Hospital, Department of Pharmacy, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
| | - Guotao Tang
- Institute of Pharmacy and Pharmacology, Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| |
Collapse
|
6
|
Farha M, Nallandhighal S, Vince R, Cotta B, Stangl-Kremser J, Triner D, Morgan TM, Palapattu GS, Cieslik M, Vaishampayan U, Udager AM, Salami SS. Analysis of the Tumor Immune Microenvironment (TIME) in Clear Cell Renal Cell Carcinoma (ccRCC) Reveals an M0 Macrophage-Enriched Subtype: An Exploration of Prognostic and Biological Characteristics of This Immune Phenotype. Cancers (Basel) 2023; 15:5530. [PMID: 38067234 PMCID: PMC10705373 DOI: 10.3390/cancers15235530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 10/31/2023] [Accepted: 11/16/2023] [Indexed: 02/12/2024] Open
Abstract
There is a need to optimize the treatment of clear cell renal cell carcinoma (ccRCC) patients at high recurrence risk after nephrectomy. We sought to elucidate the tumor immune microenvironment (TIME) of localized ccRCC and understand the prognostic and predictive characteristics of certain features. The discovery cohort was clinically localized patients in the TCGA-Kidney Renal Clear Cell Carcinoma (KIRC) project (n = 382). We identified an M0 macrophage-enriched cluster (n = 25) in the TCGA-KIRC cohort. This cluster's median progression-free survival (PFS) and overall survival (OS) were 40.4 and 45.3 months, respectively, but this was not reached in the others (p = 0.0003 and <0.0001, respectively). Gene set enrichment (GSEA) analysis revealed an enrichment of epithelial to mesenchymal transition and cell cycle progression genes within this cluster, and these patients also had a lower predicted response to immune checkpoint blockade (ICB) (4% vs. 20-34%). An M0-enriched cluster (n = 9) with shorter PFS (p = 0.0006) was also identified in the Clinical Proteomics Tumor Analysis Consortium (CPTAC) cohort (n = 94). Through this characterization of the TIME in ccRCC, a cluster of patients defined by enrichment in M0 macrophages was identified that demonstrated poor prognosis and lower predicted ICB response. Pending further validation, this signature can identify localized ccRCC patients at high risk of recurrence after nephrectomy and who may require therapeutic approaches beyond ICB monotherapy.
Collapse
Affiliation(s)
- Mark Farha
- Department of Medical Education, University of Michigan Medical School, Ann Arbor, MI 48109, USA; (M.F.); (U.V.)
| | | | - Randy Vince
- Department of Urology, Michigan Medicine, Ann Arbor, MI 48109, USA
| | - Brittney Cotta
- Department of Urology, Michigan Medicine, Ann Arbor, MI 48109, USA
| | - Judith Stangl-Kremser
- Department of Urology, Michigan Medicine, Ann Arbor, MI 48109, USA
- Department of Urology, Medical University of Vienna, 1090 Vienna, Austria
| | - Daniel Triner
- Department of Urology, Michigan Medicine, Ann Arbor, MI 48109, USA
| | - Todd M. Morgan
- Department of Urology, Michigan Medicine, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA; (M.C.); (A.M.U.)
| | - Ganesh S. Palapattu
- Department of Medical Education, University of Michigan Medical School, Ann Arbor, MI 48109, USA; (M.F.); (U.V.)
- Department of Urology, Michigan Medicine, Ann Arbor, MI 48109, USA
- Department of Urology, Medical University of Vienna, 1090 Vienna, Austria
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA; (M.C.); (A.M.U.)
| | - Marcin Cieslik
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA; (M.C.); (A.M.U.)
- Department of Pathology, Michigan Medicine, Ann Arbor, MI 48109, USA
- Michigan Center for Translational Pathology, Michigan Medicine, Ann Arbor, MI 48109, USA
| | - Ulka Vaishampayan
- Department of Medical Education, University of Michigan Medical School, Ann Arbor, MI 48109, USA; (M.F.); (U.V.)
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA; (M.C.); (A.M.U.)
- Department of Medicine, Michigan Medicine, Ann Arbor, MI 48109, USA
| | - Aaron M. Udager
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA; (M.C.); (A.M.U.)
- Department of Pathology, Michigan Medicine, Ann Arbor, MI 48109, USA
- Michigan Center for Translational Pathology, Michigan Medicine, Ann Arbor, MI 48109, USA
| | - Simpa S. Salami
- Department of Medical Education, University of Michigan Medical School, Ann Arbor, MI 48109, USA; (M.F.); (U.V.)
- Department of Urology, Michigan Medicine, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA; (M.C.); (A.M.U.)
- Michigan Center for Translational Pathology, Michigan Medicine, Ann Arbor, MI 48109, USA
| |
Collapse
|
7
|
Wesolowski L, Ge J, Castillon L, Sesia D, Dyas A, Hirosue S, Caraffini V, Warren AY, Rodrigues P, Ciriello G, Patel SA, Vanharanta S. The SWI/SNF complex member SMARCB1 supports lineage fidelity in kidney cancer. iScience 2023; 26:107360. [PMID: 37554444 PMCID: PMC10405256 DOI: 10.1016/j.isci.2023.107360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 05/22/2023] [Accepted: 07/07/2023] [Indexed: 08/10/2023] Open
Abstract
Lineage switching can induce therapy resistance in cancer. Yet, how lineage fidelity is maintained and how it can be lost remain poorly understood. Here, we have used CRISPR-Cas9-based genetic screening to demonstrate that loss of SMARCB1, a member of the SWI/SNF chromatin remodeling complex, can confer an advantage to clear cell renal cell carcinoma (ccRCC) cells upon inhibition of the renal lineage factor PAX8. Lineage factor inhibition-resistant ccRCC cells formed tumors with morphological features, but not molecular markers, of neuroendocrine differentiation. SMARCB1 inactivation led to large-scale loss of kidney-specific epigenetic programs and restoration of proliferative capacity through the adoption of new dependencies on factors that represent rare essential genes across different cancers. We further developed an analytical approach to systematically characterize lineage fidelity using large-scale CRISPR-Cas9 data. An understanding of the rules that govern lineage switching could aid the development of more durable lineage factor-targeted and other cancer therapies.
Collapse
Affiliation(s)
- Ludovic Wesolowski
- MRC Cancer Unit, University of Cambridge, Hutchison/MRC Research Centre, Cambridge Biomedical Campus, Box 197, Cambridge CB2 0XZ, UK
| | - Jianfeng Ge
- MRC Cancer Unit, University of Cambridge, Hutchison/MRC Research Centre, Cambridge Biomedical Campus, Box 197, Cambridge CB2 0XZ, UK
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge CB2 0XZ, UK
| | - Leticia Castillon
- Translational Cancer Medicine Program, Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, 00014 Helsinki, Finland
| | - Debora Sesia
- Department of Computational Biology, University of Lausanne (UNIL), 1015 Lausanne, Switzerland
- Swiss Cancer Center Leman, Lausanne, Switzerland
| | - Anna Dyas
- MRC Cancer Unit, University of Cambridge, Hutchison/MRC Research Centre, Cambridge Biomedical Campus, Box 197, Cambridge CB2 0XZ, UK
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge CB2 0XZ, UK
| | - Shoko Hirosue
- MRC Cancer Unit, University of Cambridge, Hutchison/MRC Research Centre, Cambridge Biomedical Campus, Box 197, Cambridge CB2 0XZ, UK
| | - Veronica Caraffini
- MRC Cancer Unit, University of Cambridge, Hutchison/MRC Research Centre, Cambridge Biomedical Campus, Box 197, Cambridge CB2 0XZ, UK
| | - Anne Y. Warren
- Department of Histopathology, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UK
| | - Paulo Rodrigues
- MRC Cancer Unit, University of Cambridge, Hutchison/MRC Research Centre, Cambridge Biomedical Campus, Box 197, Cambridge CB2 0XZ, UK
| | - Giovanni Ciriello
- Department of Computational Biology, University of Lausanne (UNIL), 1015 Lausanne, Switzerland
- Swiss Cancer Center Leman, Lausanne, Switzerland
| | - Saroor A. Patel
- MRC Cancer Unit, University of Cambridge, Hutchison/MRC Research Centre, Cambridge Biomedical Campus, Box 197, Cambridge CB2 0XZ, UK
| | - Sakari Vanharanta
- MRC Cancer Unit, University of Cambridge, Hutchison/MRC Research Centre, Cambridge Biomedical Campus, Box 197, Cambridge CB2 0XZ, UK
- Translational Cancer Medicine Program, Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, 00014 Helsinki, Finland
- Department of Biochemistry and Developmental Biology, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland
| |
Collapse
|
8
|
Tang C, Xie AX, Liu EM, Kuo F, Kim M, DiNatale RG, Golkaram M, Chen YB, Gupta S, Motzer RJ, Russo P, Coleman J, Carlo MI, Voss MH, Kotecha RR, Lee CH, Tansey W, Schultz N, Hakimi AA, Reznik E. Immunometabolic coevolution defines unique microenvironmental niches in ccRCC. Cell Metab 2023; 35:1424-1440.e5. [PMID: 37413991 PMCID: PMC10603615 DOI: 10.1016/j.cmet.2023.06.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 03/10/2023] [Accepted: 06/06/2023] [Indexed: 07/08/2023]
Abstract
Tumor cell phenotypes and anti-tumor immune responses are shaped by local metabolite availability, but intratumoral metabolite heterogeneity (IMH) and its phenotypic consequences remain poorly understood. To study IMH, we profiled tumor/normal regions from clear cell renal cell carcinoma (ccRCC) patients. A common pattern of IMH transcended all patients, characterized by correlated fluctuations in the abundance of metabolites and processes associated with ferroptosis. Analysis of intratumoral metabolite-RNA covariation revealed that the immune composition of the microenvironment, especially the abundance of myeloid cells, drove intratumoral metabolite variation. Motivated by the strength of RNA-metabolite covariation and the clinical significance of RNA biomarkers in ccRCC, we inferred metabolomic profiles from the RNA sequencing data of ccRCC patients enrolled in 7 clinical trials, and we ultimately identifyied metabolite biomarkers associated with response to anti-angiogenic agents. Local metabolic phenotypes, therefore, emerge in tandem with the immune microenvironment, influence ongoing tumor evolution, and are associated with therapeutic sensitivity.
Collapse
Affiliation(s)
- Cerise Tang
- Computational Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Physiology, Biophysics and Systems Biology Graduate Program, Weill Cornell Medicine, New York, NY, USA
| | - Amy X Xie
- Computational Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Biochemistry, Structural Biology, Cell Biology, Developmental Biology and Molecular Biology Graduate Program, Weill Cornell Medicine, New York, NY, USA
| | - Eric Minwei Liu
- Computational Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Fengshen Kuo
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Minsoo Kim
- Computational Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Renzo G DiNatale
- Computational Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mahdi Golkaram
- Illumina, Inc., 5200 Illumina Way, San Diego, CA 92122, USA
| | - Ying-Bei Chen
- Department of Pathology, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sounak Gupta
- Department of Pathology, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Robert J Motzer
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Paul Russo
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Jonathan Coleman
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Maria I Carlo
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Martin H Voss
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ritesh R Kotecha
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Chung-Han Lee
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Wesley Tansey
- Computational Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nikolaus Schultz
- Computational Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - A Ari Hakimi
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| | - Ed Reznik
- Computational Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| |
Collapse
|
9
|
Ueda K, Ogasawara N, Ito N, Ohnishi S, Suekane H, Kurose H, Hiroshige T, Chikui K, Uemura K, Nishihara K, Nakiri M, Suekane S, Igawa T. Prognostic Value of Absolute Lymphocyte Count in Patients with Advanced Renal Cell Carcinoma Treated with Nivolumab Plus Ipilimumab. J Clin Med 2023; 12:jcm12062417. [PMID: 36983417 PMCID: PMC10053370 DOI: 10.3390/jcm12062417] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/13/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
Nivolumab and ipilimumab (NIVO + IPI) is standard therapy for patients with advanced renal cell carcinoma (RCC). Absolute lymphocyte count (ALC) is a valuable prognostic factor in patients with various cancers treated with immune checkpoint inhibitors. Herein, we determined the prognostic value of pretreatment ALC in advanced RCC patients treated with NIVO + IPI as first-line therapy. Data from 46 advanced RCC patients treated with NIVO + IPI between September 2018 and August 2022 were retrospectively reviewed and analyzed. Median progression-free survival (PFS) and overall survival (OS) were significantly shorter in patients with low than high ALC (PFS: p = 0.0095; OS: p = 0.0182). Multivariate analysis suggested that prior nephrectomy [hazard ratio (HR) = 3.854, 95% confidence interval (CI) = 1.433-10.359, p = 0.0075] and pretreatment ALC (HR = 2.513, 95% CI = 1.119-5.648, p = 0.0257) were independent factors for PFS. Our new prognostic ALNx model based on ALC and prior nephrectomy suggested that the poor-risk group was a predictor of significantly worse PFS (p < 0.0001) and OS (p = 0.0016). Collectively, the developed ALNx model may be a novel predictor of response in advanced RCC patients treated with NIVO + IPI.
Collapse
Affiliation(s)
- Kosuke Ueda
- Department of Urology, Kurume University School of Medicine, Kurume 830-0011, Japan
| | - Naoyuki Ogasawara
- Department of Urology, Kurume University School of Medicine, Kurume 830-0011, Japan
| | - Naoki Ito
- Department of Urology, Kurume University School of Medicine, Kurume 830-0011, Japan
| | - Satoshi Ohnishi
- Department of Urology, Kurume University School of Medicine, Kurume 830-0011, Japan
| | - Hiroki Suekane
- Department of Urology, Kurume University School of Medicine, Kurume 830-0011, Japan
| | - Hirofumi Kurose
- Department of Urology, Kurume University School of Medicine, Kurume 830-0011, Japan
| | - Tasuku Hiroshige
- Department of Urology, Kurume University School of Medicine, Kurume 830-0011, Japan
| | - Katsuaki Chikui
- Department of Urology, Kurume University School of Medicine, Kurume 830-0011, Japan
| | - Keiichiro Uemura
- Department of Urology, Kurume University School of Medicine, Kurume 830-0011, Japan
| | - Kiyoaki Nishihara
- Department of Urology, Kurume University School of Medicine, Kurume 830-0011, Japan
| | - Makoto Nakiri
- Department of Urology, Kurume University School of Medicine, Kurume 830-0011, Japan
| | - Shigetaka Suekane
- Department of Urology, Kurume University School of Medicine, Kurume 830-0011, Japan
| | - Tsukasa Igawa
- Department of Urology, Kurume University School of Medicine, Kurume 830-0011, Japan
| |
Collapse
|
10
|
Current and Future Biomarkers in the Management of Renal Cell Carcinoma. Urol Clin North Am 2023; 50:151-159. [DOI: 10.1016/j.ucl.2022.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
11
|
Wang N, Hakimi AA, Chen Y. XL-ing at Induction of Apoptosis in Kidney Cancer through Inhibition of BCL-XL. Clin Cancer Res 2022; 28:4600-4602. [PMID: 35984355 PMCID: PMC9633424 DOI: 10.1158/1078-0432.ccr-22-2104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/02/2022] [Accepted: 08/09/2022] [Indexed: 01/24/2023]
Abstract
Through analysis of the cancer dependency map of CRISPR and short hairpin RNA datasets, the antiapoptotic BCL-XL was found to be a selective dependency in kidney cancer. Among kidney cancers, BCL-XL inhibition is most active in those with a mesenchymal gene signature, which portends a poor prognosis and response to current therapies. See related article by Grubb et al., p. 4689.
Collapse
Affiliation(s)
- Naitao Wang
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - A. Ari Hakimi
- Department of Surgery, Urology Service, Memorial Sloan Kettering Cancer Center, New York NY 10065, USA,Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Yu Chen
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA,Department of Medicine, Genitourinary Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA,Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| |
Collapse
|