51
|
Hiltbrunner S, Fleischmann Z, Sokol ES, Zoche M, Felley-Bosco E, Curioni-Fontecedro A. Genomic landscape of pleural and peritoneal mesothelioma tumours. Br J Cancer 2022; 127:1997-2005. [PMID: 36138075 PMCID: PMC9681755 DOI: 10.1038/s41416-022-01979-0] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 08/25/2022] [Accepted: 08/31/2022] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Malignant pleural and peritoneal mesotheliomas are rare malignancies with unacceptable poor prognoses and limited treatment options. The genomic landscape is mainly characterised by the loss of tumour suppressor genes and mutations in DNA repair genes. Currently, data from next-generation sequencing (NGS) of mesothelioma tumours is restricted to a limited number of cases; moreover, data comparing molecular features of mesothelioma from the pleural and peritoneal origin with NGS are lacking. METHODS We analysed 1113 pleural mesothelioma and 355 peritoneal mesothelioma samples. All tumours were sequenced with the FoundationOne® or FoundationOne®CDx assay for detection of substitutions, insertion-deletions, copy-number alterations and selected rearrangements in at least 324 cancer genes. RESULTS This analysis revealed alterations in 19 genes with an overall prevalence of at least 2%. Alterations in BAP1, CDKN2A, CDKN2B, NF2, MTAP, TP53 and SETD2 occurred with a prevalence of at least 10%. Peritoneal, compared to pleural mesothelioma, was characterised by a lower prevalence of alterations in CDKN2A, CDKN2B and MTAP. Moreover, we could define four distinct subgroups according to alterations in BAP1 and CDKN2A/B. Alterations in Hedgehog pathway-related genes (PTCH1/2 and SUFU) and Hippo pathway-related gene (NF2) as well as KRAS, EGFR, PDGFRA/B, ERBB2 and FGFR3 were detected in both cohorts. CONCLUSION Here, we report the molecular aberrations from the largest cohort of patients with mesothelioma. This analysis identified a proportion of patients with targetable alterations and suggests that molecular profiling can identify new treatment options for patients with mesothelioma.
Collapse
Affiliation(s)
- Stefanie Hiltbrunner
- grid.412004.30000 0004 0478 9977Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland ,grid.7400.30000 0004 1937 0650University of Zurich, Zurich, Switzerland
| | - Zoe Fleischmann
- grid.418158.10000 0004 0534 4718Foundation Medicine, Cambridge, MA USA
| | - Ethan S. Sokol
- grid.418158.10000 0004 0534 4718Foundation Medicine, Cambridge, MA USA
| | - Martin Zoche
- grid.412004.30000 0004 0478 9977Pathology Department, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Emanuela Felley-Bosco
- grid.7400.30000 0004 1937 0650University of Zurich, Zurich, Switzerland ,grid.412004.30000 0004 0478 9977Laboratory of Molecular Oncology, Department of Thoracic Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Alessandra Curioni-Fontecedro
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland. .,University of Zurich, Zurich, Switzerland.
| |
Collapse
|
52
|
Bredel M, Kim H, Bonner JA. An ErbB Lineage Co-Regulon Harbors Potentially Co-Druggable Targets for Multimodal Precision Therapy in Head and Neck Squamous Cell Carcinoma. Int J Mol Sci 2022; 23:ijms232113497. [PMID: 36362284 PMCID: PMC9658814 DOI: 10.3390/ijms232113497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 10/26/2022] [Accepted: 10/26/2022] [Indexed: 11/06/2022] Open
Abstract
The ErbB lineage of oncogenic receptor tyrosine kinases is frequently overexpressed in head and neck squamous cell carcinomas. A common co-regulon triggered by the ErbB proteins; involving shared signaling circuitries; may harbor co-druggable targets or response biomarkers for potential future multimodal precision therapy in ErbB-driven head and neck squamous cell carcinoma. We here present a cohort-based; genome-wide analysis of 488 head and neck squamous cell carcinomas curated as part of The Cancer Genome Atlas Project to characterize genes that are significantly positively co-regulated with the four ErbB proteins and those that are shared among all ErbBs denoting a common ErbB co-regulon. Significant positive gene correlations involved hundreds of genes that were co-expressed with the four ErbB family members (q < 0.05). A common; overlapping co-regulon consisted of a core set of 268 genes that were uniformly co-regulated with all four ErbB genes and highly enriched for functions in chromatin organization and histone modifications. This high-priority set of genes contained ten putative antineoplastic drug-gene interactions. The nature and directionality of these ten drug-gene associations was an inhibiting interaction for seven (PIK3CB; PIK3C2B; HDAC4; FRK; PRKCE; EPHA4; and DYRK1A) of them in which the drug decreases the biological activity or expression of the gene target. For three (CHD4; ARID1A; and PBRM1) of the associations; the directionality of the interaction was such that the gene predicted sensitivit y to the drug suggesting utility as potential response biomarkers. Drug-gene interactions that predicted the gene product to be reduced by the drug included a variety of potential targeted molecular agent classes. This unbiased genome-wide analysis identified a target-rich environment for multimodal therapeutic approaches in tumors that are putatively ErbB-driven. The results of this study require preclinical validation before ultimately devising lines of combinatorial treatment strategies for ErbB-dependent head and neck squamous cell carcinomas that incorporate these findings.
Collapse
Affiliation(s)
- Markus Bredel
- Department of Radiation Oncology, O’Neal Comprehensive Cancer Center, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Correspondence: (M.B.); (J.A.B.)
| | - Hyunsoo Kim
- Lineberger Comprehensive Cancer Center, University of Northern Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - James A. Bonner
- Department of Radiation Oncology, O’Neal Comprehensive Cancer Center, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Correspondence: (M.B.); (J.A.B.)
| |
Collapse
|
53
|
Langbein LE, El Hajjar R, Kim WY, Yang H. The convergence of tumor suppressors on the type I interferon pathway in clear cell renal cell carcinoma and its therapeutic implications. Am J Physiol Cell Physiol 2022; 323:C1417-C1429. [PMID: 36154696 PMCID: PMC9662805 DOI: 10.1152/ajpcell.00255.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 08/29/2022] [Accepted: 09/17/2022] [Indexed: 01/31/2023]
Abstract
In clear cell renal cell carcinoma (ccRCC), the von Hippel-Lindau tumor suppressor gene/hypoxia inducible factor (VHL/HIF) axis lays the groundwork for tumorigenesis and is the target of many therapeutic agents. HIF activation alone, however, is largely insufficient for kidney tumor development, and secondary mutations in PBRM1, BAP1, SETD2, KDM5C, or other tumor suppressor genes are strong enablers of tumorigenesis. Interestingly, it has been discovered that VHL loss and subsequent HIF activation results in upregulation of a negative feedback loop mediated by ISGF3, a transcription factor activated by type I interferon (IFN). Secondary mutations in the aforementioned tumor suppressor genes all partially disable this negative feedback loop to facilitate tumor growth. The convergence of several cancer genes on this pathway suggests that it plays an important role in ccRCC development and maintenance. Tumors with secondary mutations that dampen the negative feedback loop may be exquisitely sensitive to its reactivation, and pharmacological activation of ISGF3 either alone or in combination with other therapies could be an effective method to treat patients with ccRCC. In this review, we examine the relevance of the type I IFN pathway to ccRCC, synthesize our current knowledge of the ccRCC tumor suppressors in its regulation, and explore how this may impact the future treatment of patients with ccRCC.
Collapse
Affiliation(s)
- Lauren E Langbein
- Department of Pathology, Anatomy, & Cell Biology, Thomas Jefferson University, Philadelphia, Pennsylvania
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Rayan El Hajjar
- Department of Pathology, Anatomy, & Cell Biology, Thomas Jefferson University, Philadelphia, Pennsylvania
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - William Y Kim
- Department of Pathology, Anatomy, & Cell Biology, Thomas Jefferson University, Philadelphia, Pennsylvania
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Haifeng Yang
- Department of Pathology, Anatomy, & Cell Biology, Thomas Jefferson University, Philadelphia, Pennsylvania
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| |
Collapse
|
54
|
Li Y, Goldberg EM, Chen X, Xu X, McGuire JT, Leuzzi G, Karagiannis D, Tate T, Farhangdoost N, Horth C, Dai E, Li Z, Zhang Z, Izar B, Que J, Ciccia A, Majewski J, Yoon AJ, Ailles L, Mendelsohn CL, Lu C. Histone methylation antagonism drives tumor immune evasion in squamous cell carcinomas. Mol Cell 2022; 82:3901-3918.e7. [PMID: 36206767 PMCID: PMC9588679 DOI: 10.1016/j.molcel.2022.09.007] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 07/27/2022] [Accepted: 09/07/2022] [Indexed: 11/06/2022]
Abstract
How cancer-associated chromatin abnormalities shape tumor-immune interaction remains incompletely understood. Recent studies have linked DNA hypomethylation and de-repression of retrotransposons to anti-tumor immunity through the induction of interferon response. Here, we report that inactivation of the histone H3K36 methyltransferase NSD1, which is frequently found in squamous cell carcinomas (SCCs) and induces DNA hypomethylation, unexpectedly results in diminished tumor immune infiltration. In syngeneic and genetically engineered mouse models of head and neck SCCs, NSD1-deficient tumors exhibit immune exclusion and reduced interferon response despite high retrotransposon expression. Mechanistically, NSD1 loss results in silencing of innate immunity genes, including the type III interferon receptor IFNLR1, through depletion of H3K36 di-methylation (H3K36me2) and gain of H3K27 tri-methylation (H3K27me3). Inhibition of EZH2 restores immune infiltration and impairs the growth of Nsd1-mutant tumors. Thus, our work uncovers a druggable chromatin cross talk that regulates the viral mimicry response and enables immune evasion of DNA hypomethylated tumors.
Collapse
Affiliation(s)
- Yinglu Li
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Elizabeth M Goldberg
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Xiao Chen
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Xinjing Xu
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - John T McGuire
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Giuseppe Leuzzi
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Dimitris Karagiannis
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Tiffany Tate
- Department of Urology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Nargess Farhangdoost
- Department of Human Genetics, McGill University, Montreal, QC H3A 1B1, Canada; McGill University Genome Centre, Montreal, QC H3A 0G1, Canada
| | - Cynthia Horth
- Department of Human Genetics, McGill University, Montreal, QC H3A 1B1, Canada; McGill University Genome Centre, Montreal, QC H3A 0G1, Canada
| | - Esther Dai
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Zhiming Li
- Institute for Cancer Genetics, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Zhiguo Zhang
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA; Institute for Cancer Genetics, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Pediatrics, Columbia University Irving Medical Center, New York, NY 10032, USA; Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Benjamin Izar
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Medicine, Division of Hematology and Oncology, Columbia University Irving Medical Center, New York, NY 10032, USA; Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Jianwen Que
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia Center for Human Development, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Alberto Ciccia
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA; Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Jacek Majewski
- Department of Human Genetics, McGill University, Montreal, QC H3A 1B1, Canada; McGill University Genome Centre, Montreal, QC H3A 0G1, Canada
| | - Angela J Yoon
- Division of Oral and Maxillofacial Pathology, Columbia University College of Dental Medicine and Department of Pathology & Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Laurie Ailles
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Cathy Lee Mendelsohn
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Urology, Columbia University Irving Medical Center, New York, NY 10032, USA; Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Chao Lu
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA; Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY 10032, USA.
| |
Collapse
|
55
|
Zhang L, Sun T, Wu XY, Fei FM, Gao ZZ. Delineation of a SMARCA4-specific competing endogenous RNA network and its function in hepatocellular carcinoma. World J Clin Cases 2022; 10:10501-10515. [PMID: 36312469 PMCID: PMC9602240 DOI: 10.12998/wjcc.v10.i29.10501] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 08/14/2022] [Accepted: 08/30/2022] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a common malignancy worldwide, and the mortality rate continues to rise each year. SMARCA4 expression has been associated with poor prognosis in various types of cancer; however, the specific mechanism of action of SMARCA4 in HCC needs to be fully elucidated.
AIM To explore the specific mechanism of action of SMARCA4 in HCC.
METHODS Herein, the expression level of SMARCA4 as well as its association with HCC prognosis were evaluated using transcriptome profiling and clinical data of 18 different types of cancer collected from The Cancer Genome Atlas database. Furthermore, SMARCA4-high and -low groups were identified. Thereafter, gene ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analyses were performed to identify the function of SMARCA4, followed by construction of a SMARCA4-specific competing endogenous RNA (ceRNA) network using starBase database. The role of SMARCA4 in immunotherapy and its association with immune cells were assessed using correlation analysis.
RESULTS It was observed that SMARCA4 was overexpressed and negatively correlated with prognosis in HCC. Further, SMARCA4 expression was positively associated with tumor mutational burden, microsatellite stability, and immunotherapy efficacy. The SNHG3/THUMP3-AS1-miR-139-5p-SMARCA4 ceRNA network was established and could be assumed to serve as a stimulatory mechanism in HCC.
CONCLUSION The findings of this study demonstrated that SMARCA4 plays a significant role in progression and immune infiltration in HCC. Moreover, a ceRNA network was detected, which was found to be correlated with poor prognosis in HCC. The findings of this study could contribute towards the identification of predictive markers for immunotherapy and a novel mechanism of action for HCC treatment.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Clinical Oncology, Jiaxing Second Hospital, Jiaxing 314000, Zhejiang Province, China
| | - Ting Sun
- Department of Clinical Oncology, Jiaxing Second Hospital, Jiaxing 314000, Zhejiang Province, China
| | - Xiao-Ye Wu
- Department of Clinical Oncology, Jiaxing Second Hospital, Jiaxing 314000, Zhejiang Province, China
| | - Fa-Ming Fei
- Department of Clinical Oncology, The Second Affiliated Hospital of Jiaxing University, Jiaxing 314000, Zhejiang Province, China
| | - Zhen-Zhen Gao
- Department of Clinical Oncology, The Second Affiliated Hospital of Jiaxing University, Jiaxing 314000, Zhejiang Province, China
| |
Collapse
|
56
|
Rappold PM, Vuong L, Leibold J, Chakiryan NH, Curry M, Kuo F, Sabio E, Jiang H, Nixon BG, Liu M, Berglund AE, Silagy AW, Mascareno A, Golkaram M, Marker M, Reising A, Savchenko A, Millholland J, Chen YB, Russo P, Coleman J, Reznik E, Manley BJ, Ostrovnaya I, Makarov V, DiNatale RG, Blum KA, Ma X, Chowell D, Li MO, Solit DB, Lowe SW, Chan TA, Motzer RJ, Voss MH, Hakimi AA. A Targetable Myeloid Inflammatory State Governs Disease Recurrence in Clear-Cell Renal Cell Carcinoma. Cancer Discov 2022; 12:2308-2329. [PMID: 35758895 PMCID: PMC9720541 DOI: 10.1158/2159-8290.cd-21-0925] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 04/22/2022] [Accepted: 06/22/2022] [Indexed: 11/16/2022]
Abstract
It is poorly understood how the tumor immune microenvironment influences disease recurrence in localized clear-cell renal cell carcinoma (ccRCC). Here we performed whole-transcriptomic profiling of 236 tumors from patients assigned to the placebo-only arm of a randomized, adjuvant clinical trial for high-risk localized ccRCC. Unbiased pathway analysis identified myeloid-derived IL6 as a key mediator. Furthermore, a novel myeloid gene signature strongly correlated with disease recurrence and overall survival on uni- and multivariate analyses and is linked to TP53 inactivation across multiple data sets. Strikingly, effector T-cell gene signatures, infiltration patterns, and exhaustion markers were not associated with disease recurrence. Targeting immunosuppressive myeloid inflammation with an adenosine A2A receptor antagonist in a novel, immunocompetent, Tp53-inactivated mouse model significantly reduced metastatic development. Our findings suggest that myeloid inflammation promotes disease recurrence in ccRCC and is targetable as well as provide a potential biomarker-based framework for the design of future immuno-oncology trials in ccRCC. SIGNIFICANCE Improved understanding of factors that influence metastatic development in localized ccRCC is greatly needed to aid accurate prediction of disease recurrence, clinical decision-making, and future adjuvant clinical trial design. Our analysis implicates intratumoral myeloid inflammation as a key driver of metastasis in patients and a novel immunocompetent mouse model. This article is highlighted in the In This Issue feature, p. 2221.
Collapse
Affiliation(s)
- Phillip M. Rappold
- Department of Surgery, Urology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Lynda Vuong
- Department of Surgery, Urology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, MSKCC, New York, NY, USA
| | - Josef Leibold
- Cancer Biology and Genetics Program, MSKCC, New York, NY, USA
- Department of Medical Oncology & Pneumology (Internal Medicine VIII), University Hospital Tuebingen, Tuebingen 72076, Germany
- DFG Cluster of Excellence 2180 Image-Guided and Functional Instructed Tumor Therapy (iFIT), University of Tuebingen, Tuebingen 72076, Germany
| | - Nicholas H. Chakiryan
- Department of Genitourinary Oncology, H Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Michael Curry
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Fengshen Kuo
- Department of Surgery, Urology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, MSKCC, New York, NY, USA
| | - Erich Sabio
- Human Oncology and Pathogenesis Program, MSKCC, New York, NY, USA
| | - Hui Jiang
- Department of Surgery, Urology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, MSKCC, New York, NY, USA
| | - Briana G. Nixon
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ming Liu
- Legend Biotech USA Inc, NJ, USA
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Anders E. Berglund
- Department of Biostatistics and Bioinformatics, H Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Andrew W. Silagy
- Department of Surgery, Urology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ankur Mascareno
- Department of Surgery, Urology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, MSKCC, New York, NY, USA
| | - Mahdi Golkaram
- Illumina, Inc., 5200 Illumina Way, San Diego, CA 92122, USA
| | | | | | | | | | | | - Paul Russo
- Department of Surgery, Urology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jonathan Coleman
- Department of Surgery, Urology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ed Reznik
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Brandon J. Manley
- Department of Genitourinary Oncology, H Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA Integrated Mathematical Oncology Department, H Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Irina Ostrovnaya
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Vladimir Makarov
- Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, OH, USA
| | - Renzo G. DiNatale
- Department of Surgery, Urology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kyle A. Blum
- Department of Surgery, Urology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Xiaoxiao Ma
- Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, OH, USA
| | - Diego Chowell
- Department of Oncological Sciences, The Precision Immunology Institute, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ming O. Li
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - David B. Solit
- Human Oncology and Pathogenesis Program, MSKCC, New York, NY, USA
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, MSKCC, New York, NY, USA
| | - Scott W. Lowe
- Cancer Biology and Genetics Program, MSKCC, New York, NY, USA
| | - Timothy A. Chan
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, OH, USA
| | - Robert J. Motzer
- Department of Medicine, Genitourinary Oncology, MSKCC, New York, NY, USA
| | - Martin H. Voss
- Department of Medicine, Genitourinary Oncology, MSKCC, New York, NY, USA
| | - A. Ari Hakimi
- Department of Surgery, Urology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
57
|
Roviello G, Bersanelli M, Catalano M. The latest developments in biomarkers predicting response to immunotherapy. Immunotherapy 2022; 14:1085-1088. [PMID: 35924468 DOI: 10.2217/imt-2022-0053] [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: 03/14/2022] [Accepted: 07/08/2022] [Indexed: 11/21/2022] Open
Abstract
Tweetable abstract Biomarkers predicting response to immune checkpoint inhibitors can facilitate the selection of patients who benefit from immunotherapy, avoiding rapid disease progression in nonresponders and needless toxicity.
Collapse
Affiliation(s)
- Giandomenico Roviello
- Department of Health Sciences, Section of Clinical Pharmacology & Oncology, University of Florence, Viale Pieraccini, 6, Florence, 50139, Italy
| | - Melissa Bersanelli
- Medical Oncology, University Hospital of Parma, Via Gramsci 14, Parma, 43126, Italy
| | - Martina Catalano
- School of Human Health Sciences, University of Florence, Largo Brambilla 3, Florence, 50134, Italy
| |
Collapse
|
58
|
Determining Front-Line Therapeutic Strategy for Metastatic Clear Cell Renal Cell Carcinoma. Cancers (Basel) 2022; 14:cancers14194607. [PMID: 36230530 PMCID: PMC9559659 DOI: 10.3390/cancers14194607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/15/2022] [Accepted: 09/20/2022] [Indexed: 11/17/2022] Open
Abstract
The therapeutic landscape for metastatic renal cell carcinoma has rapidly evolved over the years, and we are now in an era of combination therapy strategies employing immune checkpoint blockade and anti-angiogenesis targeted therapy. Since 2018, we have gained regulatory approval for four distinct combination therapies, all with survival benefits, and with guideline recommendation for use in the front-line setting. As such, treatment selection has become increasingly complex with a myriad of treatment choices but little high-level head-to-head data to guide treatment selection. Heterogeneity in tumor biology further complicates treatment selection as tumors vary in behavior and treatment responsiveness. Ongoing development of biomarkers will certainly assist in this setting, and validation of predictive markers represents an unmet need. In their absence, we highlight features of disease and nuances to datasets from landmark prospective clinical trials to help inform treatment selection. There is growing evidence to support deferring upfront systemic therapy in some patients, with opportunities for active surveillance or metastasis-directed therapy. In others, upfront systemic therapy is warranted and necessitates thoughtful consideration of multiple clinicopathologic parameters to inform optimal patient-centered decision making.
Collapse
|
59
|
Ding H, Wang G, Yu Z, Sun H, Wang L. Role of interferon-gamma (IFN-γ) and IFN-γ receptor 1/2 (IFNγR1/2) in regulation of immunity, infection, and cancer development: IFN-γ-dependent or independent pathway. Biomed Pharmacother 2022; 155:113683. [PMID: 36095965 DOI: 10.1016/j.biopha.2022.113683] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 08/27/2022] [Accepted: 09/07/2022] [Indexed: 11/02/2022] Open
Abstract
IFN-γ, a soluble cytokine being produced by T lymphocytes, macrophages, mucosal epithelial cells, or natural killer cells, is able to bind to the IFN-γ receptor (IFNγR) and in turn activate the Janus kinase (JAK)-signal transducer and transcription protein (STAT) pathway and induce expression of IFN-γ-stimulated genes. IFN-γ is critical for innate and adaptive immunity and aberrant IFN-γ expression and functions have been associated with different human diseases. However, the IFN-γ/IFNγR signaling could be a double-edged sword in cancer development because the tissue microenvironments could determine its anti- or pro-tumorigenic activities. The IFNγR protein consists of two IFNγR1 and IFNγR2 chains, subunits of which play different roles under certain conditions. This review assessed IFNγR polymorphisms, expression and functions in development and progression of various human diseases in an IFN-γ-dependent or independent manner. This review also discussed tumor microenvironment, microbial infection, and vital molecules in the IFN-γ upstream signaling that might regulate IFNγR expression, drug resistance, and druggable strategy, to provide evidence for further application of IFNγR.
Collapse
Affiliation(s)
- Huihui Ding
- School of Pharmacy, Shandong First Medical University, Jinan, Shandong, China.
| | - Gongfu Wang
- Center for Drug Evaluation, China Food and Drug Administration (CFDA), Beijing, China.
| | - Zhen Yu
- Department of Pharmacy, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
| | - Huimin Sun
- School of Pharmacy, Shandong First Medical University, Jinan, Shandong, China.
| | - Lu Wang
- School of Pharmacy, Shandong First Medical University, Jinan, Shandong, China; Department of Pharmacy, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
| |
Collapse
|
60
|
Yuan H, Qin X, Wang J, Yang Q, Fan Y, Xu D. The cuproptosis-associated 13 gene signature as a robust predictor for outcome and response to immune- and targeted-therapies in clear cell renal cell carcinoma. Front Immunol 2022; 13:971142. [PMID: 36131921 PMCID: PMC9483097 DOI: 10.3389/fimmu.2022.971142] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 08/22/2022] [Indexed: 01/05/2023] Open
Abstract
Cuproptosis, the newly identified form of regulatory cell death (RCD), results from mitochondrial proteotoxic stress mediated by copper and FDX1. Little is known about significances of cuproptosis in oncogenesis. Here we determined clinical implications of cuproptosis in clear cell renal cell carcinoma (ccRCC). Based on the correlation and survival analyses of cuproptosis-correlated genes in TCGA ccRCC cohort, we constructed a cuproptosis-associated 13 gene signature (CuAGS-13) score system. In both TCGA training and two validation cohorts, when patients were categorized into high- and low-risk groups according to a median score as the cutoff, the CuAGS-13 high-risk group was significantly associated with shorter overall survival (OS) and/or progression-free survival (PFS) independently (P<0.001 for all). The CuAGS-13 score assessment could also predict recurrence and recurrence-free survival of patients at stage I - III with a high accuracy, which outperformed the ccAccB/ClearCode34 model, a well-established molecular predictor for ccRCC prognosis. Moreover, patients treated with immune checkpoint inhibitors (ICIs) acquired complete/partial remissions up to 3-time higher coupled with significantly longer PFS in the CuAGS-13 low- than high-risk groups in both training and validation cohorts of ccRCCs (7.2 - 14.1 vs. 2.1 - 3.0 months, P<0.001). The combination of ICI with anti-angiogenic agent Bevacizumab doubled remission rates in CuAGS-13 high-risk patients while did not improve the efficacy in the low-risk group. Further analyses showed a positive correlation between CuAGS-13 and TIDE scores. We also observed that the CuAGS-13 score assessment accurately predicted patient response to Sunitinib, and higher remission rates in the low-risk group led to longer PFS (Low- vs. high-risk, 13.9 vs. 5.8 months, P = 5.0e-12). Taken together, the CuAGS-13 score assessment serves as a robust predictor for survival, recurrence, and response to ICIs, ICI plus anti-angiogenic drugs and Sunitinib in ccRCC patients, which significantly improves patient stratifications for precision medicine of ccRCC.
Collapse
Affiliation(s)
- Huiyang Yuan
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China,*Correspondence: Huiyang Yuan, ; Yidong Fan, ; Dawei Xu,
| | - Xin Qin
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Jing Wang
- Department of Urologic Oncology, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Qingya Yang
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Yidong Fan
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China,*Correspondence: Huiyang Yuan, ; Yidong Fan, ; Dawei Xu,
| | - Dawei Xu
- Department of Medicine, Division of Hematology, Bioclinicum and Center for Molecular Medicine, Karolinska Institute and Karolinska University Hospital Solna, Stockholm, Sweden,*Correspondence: Huiyang Yuan, ; Yidong Fan, ; Dawei Xu,
| |
Collapse
|
61
|
Wu J, Lin R, Zhang L, Wei Y, Zhang R, Cai W, Hu W. LINC00887 Fosters Development of Clear Cell Renal Cell Carcinoma via Inhibiting CD8+ T Cell Immune Infiltration. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:2582474. [PMID: 36060659 PMCID: PMC9436564 DOI: 10.1155/2022/2582474] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 06/27/2022] [Indexed: 11/25/2022]
Abstract
BACKGROUND lncRNAs affect adaptive and innate immunity of cancer via mediating functional states of immune cells, genes, and pathways. Nonetheless, little is known about the molecular mechanism of lncRNA-mediated CD8+ T cell immune infiltration in progression of clear cell renal cell carcinoma (ccRCC). We designed this work to investigate the role of LINC00887 in regulating CD8+ T cell immune infiltration in ccRCC. METHODS Correlation between LINC00887 and immune factors and the expression level of LINC00887 in ccRCC were analyzed by bioinformatics methods (TCGA-KIRC database, "edgeR" package, "clusterProfiler" package, and "CIBERSORT" package). LINC00887 expression in ccRCC was examined via RT-qPCR. The cytokilling capacity of CD8+ T cells was evaluated by the lactate dehydrogenase assay. The apoptotic ability of CD8+ T cells was measured by flow cytometry. The chemotactic ability of CD8+ T cells was revealed by chemotaxis assay. CXCR3, CXCL9, and CXCL10 levels were assessed by RT-qPCR. RESULTS As suggested by bioinformatics analysis, LINC00887 was markedly upregulated in ccRCC patients and associated with expression of immune-suppression molecule, thereby abating the immune infiltration level of CD8+ cells in tumor tissue. As revealed by cellular assay, LINC00887 was upregulated in ccRCC cells, and knockdown of LINC00887 resulted in a decreased PD-L1 expression, increased CD8+ T cell toxicity, decreased apoptotic levels, and enhanced chemotaxis. Moreover, we found that LINC00887 exhibited inhibitory effect on immune infiltration of CD8+ cells in clinical tissues. CONCLUSIONS The results of this study suggested that LINC00887 promoted ccRCC progression by inhibiting immune infiltration of CD8+ T cells, providing new insights into pathogenesis of ccRCC and suggesting LINC00887 being a promising immunotherapy target for ccRCC.
Collapse
Affiliation(s)
- Jinfeng Wu
- The First School of Clinical Medicine, Southern Medical University, China
- Department of Urology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, China
| | - Rongcheng Lin
- Department of Urology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, China
| | - Lei Zhang
- The First School of Clinical Medicine, Southern Medical University, China
| | - Yongbao Wei
- Department of Urology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, China
| | - Ruochen Zhang
- Department of Urology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, China
| | - Wanghai Cai
- Department of Urology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, China
| | - Weilie Hu
- The First School of Clinical Medicine, Southern Medical University, China
- Department of Urology, Guangdong Hydropower Hospital, China
| |
Collapse
|
62
|
Rizzo M, Varnier L, Pezzicoli G, Pirovano M, Cosmai L, Porta C. IL-8 and its role as a potential biomarker of resistance to anti-angiogenic agents and immune checkpoint inhibitors in metastatic renal cell carcinoma. Front Oncol 2022; 12:990568. [PMID: 36059687 PMCID: PMC9437355 DOI: 10.3389/fonc.2022.990568] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Accepted: 08/01/2022] [Indexed: 11/24/2022] Open
Abstract
The therapeutic armamentarium of metastatic Renal Cell Carcinoma (mRCC) has consistently expanded in recent years, with the introduction of VEGF/VEGFR (Vascular Endothelial Growth Factor/Vascular Endothelial Growth Factor Receptor) inhibitors, mTOR (mammalian Target Of Rapamycin) inhibitors and Immune Checkpoint (IC) inhibitors. Currently, for the first-tline treatment of mRCC it is possible to choose between a VEGFR-TKI (VEGFR-Tyrosine Kinase Inhibitor) monotherapy, an ICI-ICI (Immune Checkpoint Inhibitor) combination and an ICI-VEGFRTKI combination. However, a consistent part of patients does not derive benefit from first-line therapy with ICIs; moreover, the use of combination regimens exposes patients to significant toxicities. Therefore, there is a critical need to develop prognostic and predictive biomarkers of response to VEGFR-TKIs and ICIs, and measurement of serum IL-8 is emerging as a potential candidate in this field. Recent retrospective analyses of large phase II and phase III trials found that elevated baseline serum IL-8 correlated with higher levels of tumor and circulating immunosuppressive myeloid cells, decreased T cell activation and poor response to treatment. These findings must be confirmed in prospective clinical trials; however, they provide evidence for a potential use of serum IL-8 as biomarker of resistance to VEGFR-TKIs and ICIs. Considering the amount of new agents and treatment regimens which are transforming the management of metastatic renal cell carcinoma, serum IL-8 could become a precious resource in tailoring the best therapy for each individual patient with the disease.
Collapse
Affiliation(s)
- Mimma Rizzo
- Division of Medical Oncology, Azienda Ospedaliero Universitaria Consorziale Policlinico di Bari, Bari, Italy
- *Correspondence: Mimma Rizzo,
| | - Luca Varnier
- Department of Pediatrics, Meyer’ Childrens University Hospital, Florence, Italy
| | - Gaetano Pezzicoli
- Department of Interdisciplinary Medicine, School of Medicine, University of Bari “A. Moro”, Bari, Italy
| | - Marta Pirovano
- Division of Nephrology and Dialysis, Azienda Socio-Sanitaria Territoriale (ASST) Fatebenefratelli-Sacco, Fatebenefratelli Hospital, Milan, Italy
| | - Laura Cosmai
- Division of Nephrology and Dialysis, Azienda Socio-Sanitaria Territoriale (ASST) Fatebenefratelli-Sacco, Fatebenefratelli Hospital, Milan, Italy
| | - Camillo Porta
- Division of Medical Oncology, Azienda Ospedaliero Universitaria Consorziale Policlinico di Bari, Bari, Italy
- Chair of Oncology, Interdisciplinary Department of Medicine, University of Bari “A. Moro”, Bari, Italy
| |
Collapse
|
63
|
Miheecheva N, Postovalova E, Lyu Y, Ramachandran A, Bagaev A, Svekolkin V, Galkin I, Zyrin V, Maximov V, Lozinsky Y, Isaev S, Ovcharov P, Shamsutdinova D, Cheng EH, Nomie K, Brown JH, Tsiper M, Ataullakhanov R, Fowler N, Hsieh JJ. Multiregional single-cell proteogenomic analysis of ccRCC reveals cytokine drivers of intratumor spatial heterogeneity. Cell Rep 2022; 40:111180. [PMID: 35977503 DOI: 10.1016/j.celrep.2022.111180] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 06/23/2022] [Accepted: 07/19/2022] [Indexed: 11/17/2022] Open
Abstract
Intratumor heterogeneity (ITH) represents a major challenge for anticancer therapies. An integrated, multidimensional, multiregional approach dissecting ITH of the clear cell renal cell carcinoma (ccRCC) tumor microenvironment (TME) is employed at the single-cell level with mass cytometry (CyTOF), multiplex immunofluorescence (MxIF), and single-nucleus RNA sequencing (snRNA-seq) and at the bulk level with whole-exome sequencing (WES), RNA-seq, and methylation profiling. Multiregional analyses reveal unexpected conservation of immune composition within each individual patient, with profound differences among patients, presenting patient-specific tumor immune microenvironment signatures despite underlying genetic heterogeneity from clonal evolution. Spatial proteogenomic TME analysis using MxIF identifies 14 distinct cellular neighborhoods and, conversely, demonstrated architectural heterogeneity among different tumor regions. Tumor-expressed cytokines are identified as key determinants of the TME and correlate with clinical outcome. Overall, this work signifies that spatial ITH occurs in ccRCC, which may drive clinical heterogeneity and warrants further interrogation to improve patient outcomes.
Collapse
Affiliation(s)
- Natalia Miheecheva
- BostonGene Corporation, University Office Park III, 95 Sawyer Road, Waltham, MA 02453, USA
| | - Ekaterina Postovalova
- BostonGene Corporation, University Office Park III, 95 Sawyer Road, Waltham, MA 02453, USA
| | - Yang Lyu
- Molecular Oncology, Division of Oncology, Department of Medicine, Washington University, St. Louis, MO 63110, USA
| | - Akshaya Ramachandran
- Molecular Oncology, Division of Oncology, Department of Medicine, Washington University, St. Louis, MO 63110, USA
| | - Alexander Bagaev
- BostonGene Corporation, University Office Park III, 95 Sawyer Road, Waltham, MA 02453, USA
| | - Viktor Svekolkin
- BostonGene Corporation, University Office Park III, 95 Sawyer Road, Waltham, MA 02453, USA
| | - Ilia Galkin
- BostonGene Corporation, University Office Park III, 95 Sawyer Road, Waltham, MA 02453, USA
| | - Vladimir Zyrin
- BostonGene Corporation, University Office Park III, 95 Sawyer Road, Waltham, MA 02453, USA
| | - Vladislav Maximov
- BostonGene Corporation, University Office Park III, 95 Sawyer Road, Waltham, MA 02453, USA
| | - Yaroslav Lozinsky
- BostonGene Corporation, University Office Park III, 95 Sawyer Road, Waltham, MA 02453, USA
| | - Sergey Isaev
- BostonGene Corporation, University Office Park III, 95 Sawyer Road, Waltham, MA 02453, USA
| | - Pavel Ovcharov
- BostonGene Corporation, University Office Park III, 95 Sawyer Road, Waltham, MA 02453, USA
| | - Diana Shamsutdinova
- BostonGene Corporation, University Office Park III, 95 Sawyer Road, Waltham, MA 02453, USA
| | - Emily H Cheng
- Human Oncology and Pathogenesis Program and Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Krystle Nomie
- BostonGene Corporation, University Office Park III, 95 Sawyer Road, Waltham, MA 02453, USA
| | - Jessica H Brown
- BostonGene Corporation, University Office Park III, 95 Sawyer Road, Waltham, MA 02453, USA
| | - Maria Tsiper
- BostonGene Corporation, University Office Park III, 95 Sawyer Road, Waltham, MA 02453, USA
| | - Ravshan Ataullakhanov
- BostonGene Corporation, University Office Park III, 95 Sawyer Road, Waltham, MA 02453, USA
| | - Nathan Fowler
- BostonGene Corporation, University Office Park III, 95 Sawyer Road, Waltham, MA 02453, USA.
| | - James J Hsieh
- Molecular Oncology, Division of Oncology, Department of Medicine, Washington University, St. Louis, MO 63110, USA.
| |
Collapse
|
64
|
The SWI/SNF chromatin remodeling factor DPF3 regulates metastasis of ccRCC by modulating TGF-β signaling. Nat Commun 2022; 13:4680. [PMID: 35945219 PMCID: PMC9363427 DOI: 10.1038/s41467-022-32472-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Accepted: 08/02/2022] [Indexed: 12/23/2022] Open
Abstract
DPF3, a component of the SWI/SNF chromatin remodeling complex, has been associated with clear cell renal cell carcinoma (ccRCC) in a genome-wide association study. However, the functional role of DPF3 in ccRCC development and progression remains unknown. In this study, we demonstrate that DPF3a, the short isoform of DPF3, promotes kidney cancer cell migration both in vitro and in vivo, consistent with the clinical observation that DPF3a is significantly upregulated in ccRCC patients with metastases. Mechanistically, DPF3a specifically interacts with SNIP1, via which it forms a complex with SMAD4 and p300 histone acetyltransferase (HAT), the major transcriptional regulators of TGF-β signaling pathway. Moreover, the binding of DPF3a releases the repressive effect of SNIP1 on p300 HAT activity, leading to the increase in local histone acetylation and the activation of cell movement related genes. Overall, our findings reveal a metastasis-promoting function of DPF3, and further establish the link between SWI/SNF components and ccRCC. The functional role of DPF3, a component of the SWI/SNF chromatin remodelling complex associated with clear cell renal cell carcinoma (ccRCC), remains unknown. Here, the authors characterise the mechanism by which DPF3 promotes metastasis via the activation of the TGF-β signalling pathway in ccRCC.
Collapse
|
65
|
Schoenfeld DA, Zhou R, Zairis S, Su W, Steinbach N, Mathur D, Bansal A, Zachem AL, Tavarez B, Hasson D, Bernstein E, Rabadan R, Parsons R. Loss of PBRM1 Alters Promoter Histone Modifications and Activates ALDH1A1 to Drive Renal Cell Carcinoma. Mol Cancer Res 2022; 20:1193-1207. [PMID: 35412614 PMCID: PMC9357026 DOI: 10.1158/1541-7786.mcr-21-1039] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 02/22/2022] [Accepted: 04/06/2022] [Indexed: 02/07/2023]
Abstract
Subunits of SWI/SNF chromatin remodeling complexes are frequently mutated in human malignancies. The PBAF complex is composed of multiple subunits, including the tumor-suppressor protein PBRM1 (BAF180), as well as ARID2 (BAF200), that are unique to this SWI/SNF complex. PBRM1 is mutated in various cancers, with a high mutation frequency in clear cell renal cell carcinoma (ccRCC). Here, we integrate RNA-seq, histone modification ChIP-seq, and ATAC-seq data to show that loss of PBRM1 results in de novo gains in H3K4me3 peaks throughout the epigenome, including activation of a retinoic acid biosynthesis and signaling gene signature. We show that one such target gene, ALDH1A1, which regulates a key step in retinoic acid biosynthesis, is consistently upregulated with PBRM1 loss in ccRCC cell lines and primary tumors, as well as non-malignant cells. We further find that ALDH1A1 increases the tumorigenic potential of ccRCC cells. Using biochemical methods, we show that ARID2 remains bound to other PBAF subunits after loss of PBRM1 and is essential for increased ALDH1A1 after loss of PBRM1, whereas other core SWI/SNF components are dispensable, including the ATPase subunit BRG1. In total, this study uses global epigenomic approaches to uncover novel mechanisms of PBRM1 tumor suppression in ccRCC. IMPLICATIONS This study implicates the SWI/SNF subunit and tumor-suppressor PBRM1 in the regulation of promoter histone modifications and retinoic acid biosynthesis and signaling pathways in ccRCC and functionally validates one such target gene, the aldehyde dehydrogenase ALDH1A1.
Collapse
Affiliation(s)
| | - Royce Zhou
- Department of Oncological Sciences, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Sakellarios Zairis
- Department of Systems Biology, Columbia University, New York, NY, 10032, USA
| | - William Su
- Department of Oncological Sciences, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Nicole Steinbach
- Department of Oncological Sciences, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Deepti Mathur
- Department of Oncological Sciences, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Ankita Bansal
- Department of Oncological Sciences, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Alexis L. Zachem
- Department of Oncological Sciences, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Bertilia Tavarez
- Department of Oncological Sciences, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Dan Hasson
- Department of Oncological Sciences, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Emily Bernstein
- Department of Oncological Sciences, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Raul Rabadan
- Department of Systems Biology, Columbia University, New York, NY, 10032, USA
| | - Ramon Parsons
- Department of Oncological Sciences, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| |
Collapse
|
66
|
Farooqi AA, Fayyaz S, Poltronieri P, Calin G, Mallardo M. Epigenetic deregulation in cancer: Enzyme players and non-coding RNAs. Semin Cancer Biol 2022; 83:197-207. [PMID: 32738290 DOI: 10.1016/j.semcancer.2020.07.013] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/26/2020] [Accepted: 07/27/2020] [Indexed: 02/08/2023]
Abstract
Data obtained from cutting-edge research have shown that deregulated epigenetic marks are critical hallmarks of cancer. Rapidly emerging scientific evidence has helped in developing a proper understanding of the mechanisms leading to control of cellular functions, from changes in chromatin accessibility, transcription and translation, and in post-translational modifications. Firstly, mechanisms of DNA methylation and demethylation are introduced, as well as modifications of DNA and RNA, with particular focus on N6-methyladenosine (m6A), discussing the effects of these modifications in normal cells and in malignancies. Then, chromatin modifying proteins and remodelling complexes are discussed. Many enzymes and accessory proteins in these complexes have been found mutated or have undergone differential splicing, leading to defective protein complexes. Epigenetic mechanisms acting on nucleosomes by polycomb repressive complexes and on chromatin by SWI/SNF complexes on nucleosome assembly/disassembly, as well as main mutated genes linked to cancers, are reviewed. Among enzymes acting on histones and other proteins erasing the reversible modifications are histone deacetylases (HDACs). Sirtuins are of interest since most of these enzymes not only deacylate histones and other proteins, but also post-translationally modify proteins adding a Mono-ADP-ribose (MAR) moiety. MAR can be read by MACRO-domain containing proteins such as histone MacroH2A1, with specific function in chromatin assembly. Finally, recent advances are presented on non-coding RNAs with a scaffold function, prospecting their role in assembly of chromatin modifying complexes, recruiting enzyme players to chromatin regions. Lastly, the imbalance in metabolites production due to mitochondrial dysfunction is presented, with the potential of these metabolites to inhibit enzymes, either writers, readers or erasers of epitranscriptome marks. In the perspectives, studies are overwied on drugs under development aiming to limit excessive enzyme activities and to reactivate chromatin modifying complexes, for therapeutic application. This knowledge may lead to novel drugs and personalised medicine for cancer patients.
Collapse
Affiliation(s)
- Ammad Ahmad Farooqi
- Department of Molecular Oncology, Institute of Biomedical and Genetic Engineering (IBGE), Islamabad 44000, Pakistan.
| | | | - Palmiro Poltronieri
- Institute of Sciences of Food Productions, National Research Council of Italy, via Monteroni Km 7, 73100 Lecce, Italy.
| | - George Calin
- Department of Experimental Therapeutics, and Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Massimo Mallardo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples, "Federico II" via Pansini 5, Napoli, Italy.
| |
Collapse
|
67
|
Feng H, Lane KA, Roumeliotis TI, Jeggo PA, Somaiah N, Choudhary JS, Downs JA. PBAF loss leads to DNA damage-induced inflammatory signaling through defective G2/M checkpoint maintenance. Genes Dev 2022; 36:gad.349249.121. [PMID: 35902118 PMCID: PMC9480851 DOI: 10.1101/gad.349249.121] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 07/13/2022] [Indexed: 11/25/2022]
Abstract
The PBRM1 subunit of the PBAF (SWI/SNF) chromatin remodeling complex is mutated in ∼40% of clear cell renal cancers. PBRM1 loss has been implicated in responses to immunotherapy in renal cancer, but the mechanism is unclear. DNA damage-induced inflammatory signaling is an important factor determining immunotherapy response. This response is kept in check by the G2/M checkpoint, which prevents progression through mitosis with unrepaired damage. We found that in the absence of PBRM1, p53-dependent p21 up-regulation is delayed after DNA damage, leading to defective transcriptional repression by the DREAM complex and premature entry into mitosis. Consequently, DNA damage-induced inflammatory signaling pathways are activated by cytosolic DNA. Notably, p53 is infrequently mutated in renal cancer, so PBRM1 mutational status is critical to G2/M checkpoint maintenance. Moreover, we found that the ability of PBRM1 deficiency to predict response to immunotherapy correlates with expression of the cytosolic DNA-sensing pathway in clinical samples. These findings have implications for therapeutic responses in renal cancer.
Collapse
Affiliation(s)
- Hugang Feng
- The Institute of Cancer Research, London SW3 6JB, United Kingdom
| | - Karen A Lane
- The Institute of Cancer Research, London SW3 6JB, United Kingdom
| | | | - Penny A Jeggo
- Genome Damage and Stability Centre, University of Sussex, Brighton BN1 9RQ, United Kingdom
| | - Navita Somaiah
- The Institute of Cancer Research, London SW3 6JB, United Kingdom
- The Royal Marsden National Health Service Foundation Trust, London SM2 5PT, United Kingdom
| | | | - Jessica A Downs
- The Institute of Cancer Research, London SW3 6JB, United Kingdom
| |
Collapse
|
68
|
Zamora-Fuentes JM, Hernández-Lemus E, Espinal-Enríquez J. Oncogenic Role of miR-217 During Clear Cell Renal Carcinoma Progression. Front Oncol 2022; 12:934711. [PMID: 35936681 PMCID: PMC9354686 DOI: 10.3389/fonc.2022.934711] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 06/13/2022] [Indexed: 11/23/2022] Open
Abstract
Clear cell renal carcinoma (ccRC) comprises a set of heterogeneous, fast-progressing pathologies with poor prognosis. Analyzing ccRC progression in terms of modifications at the molecular level may provide us with a broader understanding of the disease, paving the way for improved diagnostics and therapeutics. The role of micro-RNAs (miRs) in cancer by targeting both oncogenes and tumor suppressor genes is widely known. Despite this knowledge, the role of specific miRs and their targets in the progression of ccRC is still unknown. To evaluate the action of miRs and their target genes during ccRC progression, here we implemented a three-step method for constructing miR–gene co-expression networks for each progression stage of ccRC as well as for adjacent-normal renal tissue (NT). In the first step, we inferred all miR–gene co-expression interactions for each progression stage of ccRC and for NT. Afterwards, we filtered the whole miR–gene networks by differential gene and miR expression between successive stages: stage I with non-tumor, stage II with stage I, and so on. Finally, all miR–gene interactions whose relationships were inversely proportional (overexpressed miR and underexpressed genes and vice versa) were kept and removed otherwise. We found that miR-217 is differentially expressed in all contrasts; however, its targets were different depending on the ccRC stage. Furthermore, the target genes of miR-217 have a known role in cancer progression—for instance, in stage II network, GALNTL6 is overexpressed, and it is related to cell signaling, survival, and proliferation. In the stage III network, WNK2, a widely known tumor suppressor, is underexpressed. For the stage IV network, IGF2BP2, a post-transcriptional regulator of MYC and PTEN, is overexpressed. This data-driven network approach has allowed us to discover miRs that have different targets through ccRC progression, thus providing a method for searching possible stage-dependent therapeutic targets in this and other types of cancer.
Collapse
Affiliation(s)
| | - Enrique Hernández-Lemus
- Computational Genomics Division, National Institute of Genomic Medicine, Mexico City, Mexico
- Centro de Ciencias de la Complejidad, Universidad Nacional Autόnoma de México, Mexico City, Mexico
| | - Jesús Espinal-Enríquez
- Computational Genomics Division, National Institute of Genomic Medicine, Mexico City, Mexico
- Centro de Ciencias de la Complejidad, Universidad Nacional Autόnoma de México, Mexico City, Mexico
- *Correspondence: Jesús Espinal-Enríquez,
| |
Collapse
|
69
|
Hua S, Xie Z, Zhang Y, Wu L, Shi F, Wang X, Xia S, Dong S, Jiang J. Identification and validation of an immune-related gene prognostic signature for clear cell renal carcinoma. Front Immunol 2022; 13:869297. [PMID: 35936012 PMCID: PMC9352939 DOI: 10.3389/fimmu.2022.869297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 06/27/2022] [Indexed: 12/03/2022] Open
Abstract
Clear Cell Renal Carcinoma (ccRCC) accounts for nearly 80% of renal carcinoma cases, and immunotherapy plays an important role in ccRCC therapy. However, the responses to immunotherapy and overall survival for ccRCC patients are still hard to predict. Here, we constructed an immune-related predictive signature using 19 genes based on TCGA datasets. We also analyzed its relationships between disease prognosis, infiltrating immune cells, immune subtypes, mutation load, immune dysfunction, immune escape, etc. We found that our signature can distinguish immune characteristics and predict immunotherapeutic response for ccRCC patients with better prognostic prediction value than other immune scores. The expression levels of prognostic genes were determined by RT-qPCR assay. This signature may help to predict overall survival and guide the treatment for patients with ccRCC.
Collapse
Affiliation(s)
- Shan Hua
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhiwen Xie
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yongqing Zhang
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lei Wu
- Department of Urology, Shanghai General Hospital of Nanjing Medical University, Shanghai, China
| | - Fei Shi
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xingjie Wang
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shujie Xia
- Department of Urology, Shanghai General Hospital of Nanjing Medical University, Shanghai, China
| | - Shengli Dong
- Nursing Department, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Shengli Dong, ; Juntao Jiang,
| | - Juntao Jiang
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Shengli Dong, ; Juntao Jiang,
| |
Collapse
|
70
|
Acharya N, Singh KP. Recent advances in the molecular basis of chemotherapy resistance and potential application of epigenetic therapeutics in chemorefractory renal cell carcinoma. WIREs Mech Dis 2022; 14:e1575. [DOI: 10.1002/wsbm.1575] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 06/11/2022] [Accepted: 06/22/2022] [Indexed: 12/13/2022]
Affiliation(s)
- Narayan Acharya
- Department of Environmental Toxicology, The Institute of Environmental and Human Health (TIEHH) Texas Tech University Lubbock Texas USA
| | - Kamaleshwar P. Singh
- Department of Environmental Toxicology, The Institute of Environmental and Human Health (TIEHH) Texas Tech University Lubbock Texas USA
| |
Collapse
|
71
|
Zhou Z, Huang D, Yang S, Liang J, Wang X, Rao Q. Clinicopathological Significance, Related Molecular Changes and Tumor Immune Response Analysis of the Abnormal SWI/SNF Complex Subunit PBRM1 in Gastric Adenocarcinoma. PATHOLOGY AND ONCOLOGY RESEARCH 2022; 28:1610479. [PMID: 35928964 PMCID: PMC9344308 DOI: 10.3389/pore.2022.1610479] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 06/15/2022] [Indexed: 12/03/2022]
Abstract
Background: PBRM1 gene abnormalities were recently found to play a role in tumor development and tumor immune activity. This article will explore the clinicopathological and molecular changes and tumor immune activity of the abnormal SWI/SNF complex subunit PBRM1 in gastric adenocarcinoma (GAC) and its significance. Methods: The cBioPortal, LinkedOmics and TISIDB datasets were used to analyze the abnormality of the PBRM1 gene in GAC and its relationship with prognosis, related molecular changes and tumor-infiltrating lymphocytes (TILs). In addition, 198 GAC cases were collected to further study the relationship between the loss/attenuation of PBRM1 expression and clinicopathology, prognosis, microsatellite stability, PD-L1 expression and TIL in GAC. DNA whole-exome sequencing was performed on 7 cases of gastric cancer with loss of PBRM1 expression. Results: The cBioPortal data showed that PBRM1 deletion/mutation accounted for 7.32% of GAC and was significantly associated with several molecular changes, such as molecular subtypes of GAC. The LinkedOmics dataset showed that PBRM1 mutation and its promoter DNA methylation showed lower PBRM1 mRNA expression, and PBRM1 mutation cases showed significantly higher mRNA expression of PD-L1 (CD274). TISIDB data showed that PBRM1 abnormalities were significantly positively associated with multiple TILs. In our group of 198 cases, the loss/attenuation of PBRM1 expression was significantly positively correlated with intra-tumoral tumor infiltrating lymphocytes (iTILs) and deficient MMR and PD-L1 expression. Kaplan–Meier survival analysis showed that the overall survival of GAC patients with loss/attenuation of PBRM1 expression was significantly better (p = 0.023). iTIL was an independent prognostic factor of GAC. Loss of PBRM1 expression often co-occurs with mutations in other SWI/SNF complex subunit genes, and there are some repetitive KEGG signaling changes. Conclusion: Abnormality of the PBRM1 gene may be related to the occurrence of some GACs and can affect tumor immune activity, thereby affecting clinicopathology and prognosis. It may be a potentially effective predictive marker for immunotherapy and a novel therapeutic approach associated with synthetic lethality.
Collapse
Affiliation(s)
- Zhiyi Zhou
- Department of Pathology, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, China
| | - Dandan Huang
- Digestive Endoscopic Center, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, China
| | - Shudong Yang
- Department of Pathology, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, China
| | - Jiabei Liang
- Department of Pathology, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, China
| | - Xuan Wang
- Department of Pathology, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Qiu Rao
- Department of Pathology, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
- *Correspondence: Qiu Rao,
| |
Collapse
|
72
|
Halima A, Vuong W, Chan TA. Next-generation sequencing: unraveling genetic mechanisms that shape cancer immunotherapy efficacy. J Clin Invest 2022; 132:154945. [PMID: 35703181 PMCID: PMC9197511 DOI: 10.1172/jci154945] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Immunity is governed by fundamental genetic processes. These processes shape the nature of immune cells and set the rules that dictate the myriad complex cellular interactions that power immune systems. Everything from the generation of T cell receptors and antibodies, control of epitope presentation, and recognition of pathogens by the immunoediting of cancer cells is, in large part, made possible by core genetic mechanisms and the cellular machinery that they encode. In the last decade, next-generation sequencing has been used to dissect the complexities of cancer immunity with potent effect. Sequencing of exomes and genomes has begun to reveal how the immune system recognizes “foreign” entities and distinguishes self from non-self, especially in the setting of cancer. High-throughput analyses of transcriptomes have revealed deep insights into how the tumor microenvironment affects immunotherapy efficacy. In this Review, we discuss how high-throughput sequencing has added to our understanding of how immune systems interact with cancer cells and how cancer immunotherapies work.
Collapse
Affiliation(s)
- Ahmed Halima
- Department of Radiation Oncology, Taussig Cancer Institute, and
| | - Winston Vuong
- Department of Radiation Oncology, Taussig Cancer Institute, and
| | - Timothy A Chan
- Department of Radiation Oncology, Taussig Cancer Institute, and.,Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, Ohio, USA.,National Center for Regenerative Medicine, Cleveland, Ohio, USA
| |
Collapse
|
73
|
Molecular Subtypes Based on Genomic and Transcriptomic Features Correlate with the Responsiveness to Immune Checkpoint Inhibitors in Metastatic Clear Cell Renal Cell Carcinoma. Cancers (Basel) 2022; 14:cancers14102354. [PMID: 35625960 PMCID: PMC9139776 DOI: 10.3390/cancers14102354] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 05/04/2022] [Accepted: 05/04/2022] [Indexed: 12/29/2022] Open
Abstract
Simple Summary Immune checkpoint inhibitors (ICIs), such as programmed cell death protein 1 (PD-1) blockade, have proven to be the most effective agents for the management of many cancer types. Although ICIs are the current standard of care for treating metastatic clear cell renal cell carcinoma (ccRCC), 40–60% of patients still have intrinsic resistance to ICIs across multiple clinical trials. Therefore, identifying optimal biomarkers that can predict either responders or non-responders to ICIs has been of tremendous importance. Here, we generated targeted sequencing and whole transcriptomic sequencing of 60 patients with metastatic ccRCC treated with ICIs. Moreover, transcriptomic analysis was integrated to identify molecular subtypes using a total of 177 tumor samples by merging our data and published data derived from the CheckMate 025 trial. Our results show that these molecular subtypes are associated with specific genomic alterations, distinct molecular pathways, and differential clinical outcomes in patients with metastatic ccRCC treated with ICIs. Abstract Clear cell renal cell carcinoma (ccRCC) has been reported to be highly immune to and infiltrated by T cells and has angiogenesis features, but the effect of given features on clinical outcomes followed by immune checkpoint inhibitors (ICIs) in ccRCC has not been fully characterized. Currently, loss of function mutation in PBRM1, a PBAF-complex gene frequently mutated in ccRCC, is associated with clinical benefit from ICIs, and is considered as a predictive biomarker for response to anti-PD-1 therapy. However, functional mechanisms of PBRM1 mutation regarding immunotherapy responsiveness are still poorly understood. Here, we performed targeted sequencing (n = 60) and whole transcriptomic sequencing (WTS) (n = 61) of patients with metastatic ccRCC treated by ICIs. By integrating WTS data from the CheckMate 025 trial, we obtained WTS data of 177 tumors and finally identified three molecular subtypes that are characterized by distinct molecular phenotypes and frequency of PBRM1 mutations. Patient clustered subtypes 1 and 3 demonstrated worse responses and survival after ICIs treatment, with a low proportion of PBRM1 mutation and angiogenesis-poor, but were immune-rich and cell-cycle enriched. Notably, patients clustered in the subtype 2 showed a better response and survival after ICIs treatment, with enrichment of PBRM1 mutation and metabolic programs and a low exhausted immune phenotype. Further analysis of the subtype 2 population demonstrated that GATM (glycine amidinotransferase), as a novel gene associated with PBRM1 mutation, plays a pivotal role in ccRCC by using a cell culture model, revealing tumor, suppressive-like features in reducing proliferation and migration. In summary, we identified that metastatic ccRCC treated by ICIs have distinct genomic and transcriptomic features that may account for their responsiveness to ICIs. We also revealed that the novel gene GATM can be a potential tumor suppressor and/or can be associated with therapeutic efficacy in metastatic ccRCC treated by ICIs.
Collapse
|
74
|
Kong SK, Kim BS, Lim H, Kim HJ, Kim YS. Dissection of PD-L1 promoter reveals differential transcriptional regulation of PD-L1 in VHL mutant clear cell renal cell carcinoma. J Transl Med 2022; 102:352-362. [PMID: 34789838 DOI: 10.1038/s41374-021-00703-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 11/03/2021] [Accepted: 11/03/2021] [Indexed: 11/09/2022] Open
Abstract
Programmed death-ligand 1 (PD-L1) is constitutively expressed by hypoxia-inducible factor 2α (HIF2α). It can be induced by interferon gamma (IFNγ) signaling in clear cell renal cell carcinoma (ccRCC). Clinical trials of metastatic ccRCCs have suggested that a canonical IFNγ signature is a better biomarker for therapeutic response to immune checkpoint inhibitors (ICIs) than PD-L1 expression levels in tumor cells. To understand the therapeutic response to ICIs according to PD-L1 expression levels, we analyzed transcriptional regulation of the PD-L1 promoter by HIF2α and IFNγ-inducible interferon regulatory factor-1 (IRF-1) in ccRCC cells. Here, we present two ccRCC cell models showing differential PD-L1 expression levels in response to IFNγ and hypoxia. Analysis of The Cancer Genome Atlas RNA-sequencing data revealed that PD-L1 expression correlated with JAK2 and STAT1 expression of the canonical IFNγ signature in ccRCC tissues. Upon IFNγ stimulation, PD-L1 was induced by sequential activation of JAK2/STAT1/IRF-1 signaling in both WT- and Mut- VHL ccRCC cells. IFNγ activated the IRF-1α site of the PD-L1 promoter. The IFNγ-mediated increase of PD-L1 expression in Mut-VHL cells was 4.8-fold greater than that in WT-VHL cells. Under normoxia condition, PD-L1 expression in Mut-VHL cells was significantly higher than that in WT-VHL cells due to high basal HIF2α expression. Under hypoxia condition, PD-L1 expression in WT-VHL cells was induced up to 1.8-fold through activation of hypoxia-response elements 2 and 3. In contrast, although PD-L1 in Mut-VHL cells was already highly expressed in the basal state through activation of hypoxia-response elements 2, 3, and 4, it was no longer induced by hypoxia. In conclusion, Mut-VHL ccRCC cells displayed higher PD-L1 expression due to high basal HIF2α expression and a stronger response to IFNγ stimulation than WT-VHL cells. The fact that HIF2α antagonists can potentially reduce PD-L1 expression levels should be considered in ICI combination therapy.
Collapse
Affiliation(s)
- Su-Kang Kong
- Department of Pathology, Korea University College of Medicine, Seoul, Republic of Korea
| | - Byung Soo Kim
- Department of Pathology, Korea University College of Medicine, Seoul, Republic of Korea
| | - Hyangsoon Lim
- Department of Pathology, Korea University College of Medicine, Seoul, Republic of Korea
| | - Hyun Ji Kim
- Department of Pathology, Korea University College of Medicine, Seoul, Republic of Korea
| | - Young-Sik Kim
- Department of Pathology, Korea University College of Medicine, Seoul, Republic of Korea. .,Department of Pathology, Korea University Ansan Hospital, Ansan, Republic of Korea.
| |
Collapse
|
75
|
Jiang W, Zeng H, Liu Z, Jin K, Hu B, Chang Y, Liu L, Zhu Y, Xu L, Wang Z, Guo J, Xu J. Immune inactivation by CD47 expression predicts clinical outcomes and therapeutic responses in clear cell renal cell carcinoma patients. Urol Oncol 2022; 40:166.e15-166.e25. [PMID: 34998671 DOI: 10.1016/j.urolonc.2021.11.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 11/16/2021] [Accepted: 11/29/2021] [Indexed: 10/19/2022]
Abstract
BACKGROUND CD47 has been identified as a phagocytosis checkpoint conferring poor clinical outcomes in various cancer types. A flurry of clinical trials designed to evaluate agents that block CD47 have been initiated. We aimed to explore the clinical significance of CD47 and its correlation with immune infiltration and molecular features in clear cell renal cell carcinoma (ccRCC). METHODS 235 tumor tissue microarray specimens of ccRCC patients from Zhongshan Hospital, 530 ccRCC patients from The Cancer Genome Atlas and 726 ccRCC patients from JAVELIN Renal 101 study were analyzed. CD47 expression and immune contexture were examined by immunohistochemistry and CIBERSORT algorithm. Survival analyses were conducted through Kaplan-Meier curves and Cox regression model. RESULTS We demonstrated that ccRCC patients with high CD47 expression exhibited inferior overall survival and recurrence-free survival. CD47 expression associated with heavily immune infiltrated but immunosuppressed microenvironment. CD8+ T cells infiltration had discordant prognostic value based on CD47 expression, where high CD8+ T cell infiltration was associated with worse clinical outcome in CD47hi patients and with favorable prognosis in CD47lo patients. Patients with mutated PBRM1 and SETD2 correlated with decreased CD47 mRNA expression. Patients with higher CD47 expression possessed improved PFS in ICI + VEGFR TKI combination therapy. CONCLUSIONS CD47 expression was an independent prognosticator of clinical outcome for ccRCC patients. CD47 expression correlated with ccRCC molecular classification and response to combination therapy. The phagocytosis checkpoint CD47 could be applied as an attractive candidate for immunotherapeutic approach in ccRCC.
Collapse
Affiliation(s)
- Wenbin Jiang
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Han Zeng
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Zhaopei Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Kaifeng Jin
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Baoying Hu
- Department of Immunology, School of Medicine, Nantong University, Nantong, China
| | - Yuan Chang
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Li Liu
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yu Zhu
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Le Xu
- Department of Urology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zewei Wang
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Jianming Guo
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Jiejie Xu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China.
| |
Collapse
|
76
|
Choueiri TK, Albiges L, Atkins MB, Bakouny Z, Bratslavsky G, Braun DA, Haas NB, Haanen JB, Hakimi AA, Jewett MA, Jonasch E, Kaelin WG, Kapur P, Labaki C, Lewis B, McDermott DF, Pal SK, Pels K, Poteat S, Powles T, Rathmell WK, Rini BI, Signoretti S, Tannir NM, Uzzo RG, Hammers HJ. From Basic Science to Clinical Translation in Kidney Cancer: A Report from the Second Kidney Cancer Research Summit. Clin Cancer Res 2022; 28:831-839. [PMID: 34965942 PMCID: PMC9223120 DOI: 10.1158/1078-0432.ccr-21-3238] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 11/07/2021] [Accepted: 12/21/2021] [Indexed: 11/16/2022]
Abstract
The second Kidney Cancer Research Summit was held virtually in October 2020. The meeting gathered worldwide experts in the field of kidney cancer, including basic, translational, and clinical scientists as well as patient advocates. Novel studies were presented, addressing areas of unmet need related to different topics. These include novel metabolic targets, promising immunotherapeutic regimens, predictive genomic and transcriptomic biomarkers, and variant histologies of renal cell carcinoma (RCC). With the development of pioneering technologies, and an unprecedented commitment to kidney cancer research, the field has tremendously evolved. This perspective aims to summarize the different sessions of the conference, outline major advances in the understanding of RCC and discuss current challenges faced by the field.
Collapse
Affiliation(s)
- Toni K. Choueiri
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Laurence Albiges
- Department of Medical Oncology, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Michael B. Atkins
- Georgetown Lombardi Comprehensive Cancer Center, Washington, DC, USA
| | - Ziad Bakouny
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Gennady Bratslavsky
- Department of Urology, State University of New York (SUNY) Upstate Medical University, Syracuse, New York
| | - David A. Braun
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Naomi B. Haas
- Division of Hematology and Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - John B.A.G. Haanen
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - A Ari Hakimi
- Department of Surgery, Urology Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Michael A.S. Jewett
- Division of Urology, Department of Surgery, Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Eric Jonasch
- Department of Genitourinary Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - William G. Kaelin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Payal Kapur
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Chris Labaki
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | - David F. McDermott
- Division of Medical Oncology, Beth Israel Deaconess Medical Center, Dana-Farber/Harvard Cancer Center, Boston, Massachusetts
| | - Sumanta K. Pal
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Kevin Pels
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | - Thomas Powles
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - W. Kimryn Rathmell
- Department of Medicine, Vanderbilt University Medical Center (VUMC), Nashville, TN, USA
| | - Brian I. Rini
- Department of Medicine, Vanderbilt University Medical Center (VUMC), Nashville, TN, USA
| | - Sabina Signoretti
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Nizar M. Tannir
- Department of Genitourinary Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Robert G. Uzzo
- Department of Surgical Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Hans J. Hammers
- Division of Hematology-Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
77
|
Saiga K, Ohe C, Yoshida T, Ohsugi H, Ikeda J, Atsumi N, Noda Y, Yasukochi Y, Higasa K, Taniguchi H, Kinoshita H, Tsuta K. PBRM1 Immunohistochemical Expression Profile Correlates with Histomorphological Features and Endothelial Expression of Tumor Vasculature for Clear Cell Renal Cell Carcinoma. Cancers (Basel) 2022; 14:1062. [PMID: 35205810 PMCID: PMC8870106 DOI: 10.3390/cancers14041062] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 02/15/2022] [Accepted: 02/16/2022] [Indexed: 01/05/2023] Open
Abstract
Loss of the polybromo-1 (PBRM1) protein has been expected as a possible biomarker for clear cell renal cell carcinoma (ccRCC). There is little knowledge about how PBRM1 immunohistochemical expression correlates with the histomorphological features of ccRCC and the endothelial expression of tumor vasculature. The present study evaluates the association of architectural patterns with the PBRM1 expression of cancer cells using a cohort of 425 patients with nonmetastatic ccRCC. Furthermore, we separately assessed the PBRM1 expression of the endothelial cells and evaluated the correlation between the expression of cancer cells and endothelial cells. PBRM1 loss in cancer cells was observed in 148 (34.8%) patients. In the correlation analysis between architectural patterns and PBRM1 expression, macrocyst/microcystic, tubular/acinar, and compact/small nested were positively correlated with PBRM1 expression, whereas alveolar/large nested, thick trabecular/insular, papillary/pseudopapillary, solid sheets, and sarcomatoid/rhabdoid were negatively correlated with PBRM1 expression. PBRM1 expression in vascular endothelial cells correlated with the expression of cancer cells (correlation coefficient = 0.834, p < 0.001). PBRM1 loss in both cancer and endothelial cells was associated with a lower recurrence-free survival rate (p < 0.001). Our PBRM1 expression profile indicated that PBRM1 expression in both cancer and endothelial cells may be regulated in an orchestrated manner.
Collapse
Affiliation(s)
- Kazuho Saiga
- Department of Pathology, Kansai Medical University, 2-3-1 Shin-machi, Hirakata 573-1191, Japan; (K.S.); (J.I.); (N.A.); (Y.N.); (K.T.)
| | - Chisato Ohe
- Department of Pathology, Kansai Medical University, 2-3-1 Shin-machi, Hirakata 573-1191, Japan; (K.S.); (J.I.); (N.A.); (Y.N.); (K.T.)
| | - Takashi Yoshida
- Department of Urology and Andrology, Kansai Medical University, 2-3-1 Shin-machi, Hirakata 573-1191, Japan; (T.Y.); (H.O.); (H.T.); (H.K.)
| | - Haruyuki Ohsugi
- Department of Urology and Andrology, Kansai Medical University, 2-3-1 Shin-machi, Hirakata 573-1191, Japan; (T.Y.); (H.O.); (H.T.); (H.K.)
| | - Junichi Ikeda
- Department of Pathology, Kansai Medical University, 2-3-1 Shin-machi, Hirakata 573-1191, Japan; (K.S.); (J.I.); (N.A.); (Y.N.); (K.T.)
- Department of Urology and Andrology, Kansai Medical University, 2-3-1 Shin-machi, Hirakata 573-1191, Japan; (T.Y.); (H.O.); (H.T.); (H.K.)
| | - Naho Atsumi
- Department of Pathology, Kansai Medical University, 2-3-1 Shin-machi, Hirakata 573-1191, Japan; (K.S.); (J.I.); (N.A.); (Y.N.); (K.T.)
| | - Yuri Noda
- Department of Pathology, Kansai Medical University, 2-3-1 Shin-machi, Hirakata 573-1191, Japan; (K.S.); (J.I.); (N.A.); (Y.N.); (K.T.)
| | - Yoshiki Yasukochi
- Department of Genome Analysis, Institute of Biomedical Science, Kansai Medical University, 2-5-1 Shin-machi, Hirakata 573-1010, Japan; (Y.Y.); (K.H.)
| | - Koichiro Higasa
- Department of Genome Analysis, Institute of Biomedical Science, Kansai Medical University, 2-5-1 Shin-machi, Hirakata 573-1010, Japan; (Y.Y.); (K.H.)
| | - Hisanori Taniguchi
- Department of Urology and Andrology, Kansai Medical University, 2-3-1 Shin-machi, Hirakata 573-1191, Japan; (T.Y.); (H.O.); (H.T.); (H.K.)
| | - Hidefumi Kinoshita
- Department of Urology and Andrology, Kansai Medical University, 2-3-1 Shin-machi, Hirakata 573-1191, Japan; (T.Y.); (H.O.); (H.T.); (H.K.)
| | - Koji Tsuta
- Department of Pathology, Kansai Medical University, 2-3-1 Shin-machi, Hirakata 573-1191, Japan; (K.S.); (J.I.); (N.A.); (Y.N.); (K.T.)
| |
Collapse
|
78
|
Loeffler CML, Gaisa NT, Muti HS, van Treeck M, Echle A, Ghaffari Laleh N, Trautwein C, Heij LR, Grabsch HI, Ortiz Bruechle N, Kather JN. Predicting Mutational Status of Driver and Suppressor Genes Directly from Histopathology With Deep Learning: A Systematic Study Across 23 Solid Tumor Types. Front Genet 2022; 12:806386. [PMID: 35251119 PMCID: PMC8889144 DOI: 10.3389/fgene.2021.806386] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 12/30/2021] [Indexed: 12/12/2022] Open
Abstract
In the last four years, advances in Deep Learning technology have enabled the inference of selected mutational alterations directly from routine histopathology slides. In particular, recent studies have shown that genetic changes in clinically relevant driver genes are reflected in the histological phenotype of solid tumors and can be inferred by analysing routine Haematoxylin and Eosin (H&E) stained tissue sections with Deep Learning. However, these studies mostly focused on selected individual genes in selected tumor types. In addition, genetic changes in solid tumors primarily act by changing signaling pathways that regulate cell behaviour. In this study, we hypothesized that Deep Learning networks can be trained to directly predict alterations of genes and pathways across a spectrum of solid tumors. We manually outlined tumor tissue in H&E-stained tissue sections from 7,829 patients with 23 different tumor types from The Cancer Genome Atlas. We then trained convolutional neural networks in an end-to-end way to detect alterations in the most clinically relevant pathways or genes, directly from histology images. Using this automatic approach, we found that alterations in 12 out of 14 clinically relevant pathways and numerous single gene alterations appear to be detectable in tissue sections, many of which have not been reported before. Interestingly, we show that the prediction performance for single gene alterations is better than that for pathway alterations. Collectively, these data demonstrate the predictability of genetic alterations directly from routine cancer histology images and show that individual genes leave a stronger morphological signature than genetic pathways.
Collapse
Affiliation(s)
- Chiara Maria Lavinia Loeffler
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), Aachen, Germany
- *Correspondence: Chiara Maria Lavinia Loeffler,
| | - Nadine T. Gaisa
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), Aachen, Germany
- Institute of Pathology, University Hospital RWTH Aachen, Aachen, Germany
| | - Hannah Sophie Muti
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), Aachen, Germany
| | - Marko van Treeck
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), Aachen, Germany
| | - Amelie Echle
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), Aachen, Germany
| | - Narmin Ghaffari Laleh
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), Aachen, Germany
| | - Christian Trautwein
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), Aachen, Germany
| | - Lara R. Heij
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), Aachen, Germany
- Institute of Pathology, University Hospital RWTH Aachen, Aachen, Germany
- Department of Surgery and Transplantation, University Hospital RWTH Aachen, Aachen, Germany
- NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, Netherlands
| | - Heike I. Grabsch
- Department of Pathology, GROW School for Oncology and Reproduction, Maastricht University Medical Center+, Maastricht, Netherlands
- Pathology and Data Analytics, Leeds Institute of Medical Research at St James’s, University of Leeds, Leeds, United Kingdom
| | - Nadina Ortiz Bruechle
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), Aachen, Germany
- Institute of Pathology, University Hospital RWTH Aachen, Aachen, Germany
| | - Jakob Nikolas Kather
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), Aachen, Germany
- Pathology and Data Analytics, Leeds Institute of Medical Research at St James’s, University of Leeds, Leeds, United Kingdom
- Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
79
|
Demidova EV, Ghatalia P, Arora S. Combination Strategies for Immune Checkpoint Inhibitors in PBRM1-mutant Renal Cell Carcinoma: To PARP or Not To PARP? Eur Urol 2022; 81:149-150. [PMID: 34753637 DOI: 10.1016/j.eururo.2021.10.028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 10/20/2021] [Indexed: 11/20/2022]
Affiliation(s)
- Elena V Demidova
- Cancer Prevention and Control Program, Fox Chase Cancer Center, Philadelphia, PA, USA; Kazan Federal University, Kazan, Russia
| | - Pooja Ghatalia
- Department of Hematology/Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Sanjeevani Arora
- Cancer Prevention and Control Program, Fox Chase Cancer Center, Philadelphia, PA, USA; Department of Radiation Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA.
| |
Collapse
|
80
|
Hagiwara M, Fushimi A, Bhattacharya A, Yamashita N, Morimoto Y, Oya M, Withers HG, Hu Q, Liu T, Liu S, Wong KK, Long MD, Kufe D. MUC1-C integrates type II interferon and chromatin remodeling pathways in immunosuppression of prostate cancer. Oncoimmunology 2022; 11:2029298. [PMID: 35127252 PMCID: PMC8812775 DOI: 10.1080/2162402x.2022.2029298] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 01/10/2022] [Accepted: 01/11/2022] [Indexed: 02/06/2023] Open
Abstract
The oncogenic MUC1-C protein drives dedifferentiation of castrate resistant prostate cancer (CRPC) cells in association with chromatin remodeling. The present work demonstrates that MUC1-C is necessary for expression of IFNGR1 and activation of the type II interferon-gamma (IFN-γ) pathway. We show that MUC1-C→ARID1A/BAF signaling induces IFNGR1 transcription and that MUC1-C-induced activation of the NuRD complex suppresses FBXW7 in stabilizing the IFNGR1 protein. MUC1-C and NuRD were also necessary for expression of the downstream STAT1 and IRF1 transcription factors. We further demonstrate that MUC1-C and PBRM1/PBAF are necessary for IRF1-induced expression of (i) IDO1, WARS and PTGES, which metabolically suppress the immune tumor microenvironment (TME), and (ii) the ISG15 and SERPINB9 inhibitors of T cell function. Of translational relevance, we show that MUC1 associates with expression of IFNGR1, STAT1 and IRF1, as well as the downstream IDO1, WARS, PTGES, ISG15 and SERPINB9 immunosuppressive effectors in CRPC tumors. Analyses of scRNA-seq data further demonstrate that MUC1 correlates with cancer stem cell (CSC) and IFN gene signatures across CRPC cells. Consistent with these results, MUC1 associates with immune cell-depleted "cold" CRPC TMEs. These findings demonstrate that MUC1-C integrates chronic activation of the type II IFN-γ pathway and induction of chromatin remodeling complexes in linking the CSC state with immune evasion.
Collapse
Affiliation(s)
- Masayuki Hagiwara
- Harvard Medical School, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Atsushi Fushimi
- Harvard Medical School, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | - Nami Yamashita
- Harvard Medical School, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | - Mototsugu Oya
- Department of Urology, Keio University School of Medicine, Tokyo, Japan
| | - Henry G. Withers
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Qiang Hu
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Tao Liu
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Song Liu
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Kwok K. Wong
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Medical Center, New York, NY, USA
| | - Mark D. Long
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Donald Kufe
- Harvard Medical School, Dana-Farber Cancer Institute, Boston, MA, USA
| |
Collapse
|
81
|
Jonasch E, Atkins MB, Chowdhury S, Mainwaring P. Combination of Anti-Angiogenics and Checkpoint Inhibitors for Renal Cell Carcinoma: Is the Whole Greater Than the Sum of Its Parts? Cancers (Basel) 2022; 14:cancers14030644. [PMID: 35158916 PMCID: PMC8833428 DOI: 10.3390/cancers14030644] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/21/2022] [Accepted: 01/23/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Checkpoint inhibitors and anti-angiogenic therapies are treatments that slow the progression of renal cell carcinoma, the most common type of kidney cancer. Checkpoint inhibitors and anti-angiogenic therapies work in different ways. Checkpoint inhibitors help to prevent tumors from hiding from the body’s immune system, while anti-angiogenic therapies slow the development of blood vessels that tumours need to help them to grow. Studies have shown that treatment with combination checkpoint inhibitor plus anti-angiogenic therapy can achieve better outcomes for patients with renal cell carcinoma than treatment with anti-angiogenic therapy alone. In this review, we consider how combination checkpoint inhibitor plus anti-angiogenic therapy works, and we review the current literature to identify evidence to inform clinicians as to the most effective way to use these different types of drugs, either one after the other, or together, for maximum patient benefit. Abstract Anti-angiogenic agents, such as vascular endothelial growth factor (VEGF) receptor tyrosine kinase inhibitors and anti-VEGF antibodies, and immune checkpoint inhibitors (CPIs) are standard treatments for advanced renal cell carcinoma (aRCC). In the past, these agents were administered as sequential monotherapies. Recently, combinations of anti-angiogenic agents and CPIs have been approved for the treatment of aRCC, based on evidence that they provide superior efficacy when compared with sunitinib monotherapy. Here we explore the possible mechanisms of action of these combinations, including a review of relevant preclinical data and clinical evidence in patients with aRCC. We also ask whether the benefit is additive or synergistic, and, thus, whether concomitant administration is preferred over sequential monotherapy. Further research is needed to understand how combinations of anti-angiogenic agents with CPIs compare with CPI monotherapy or combination therapy (e.g., nivolumab and ipilimumab), and whether the long-term benefit observed in a subset of patients treated with CPI combinations will also be realised in patients treated with an anti-angiogenic therapy and a CPI. Additional research is also needed to establish whether other elements of the tumour microenvironment also need to be targeted to optimise treatment efficacy, and to identify biomarkers of response to inform personalised treatment using combination therapies.
Collapse
Affiliation(s)
- Eric Jonasch
- Department of Genitourinary Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 1374, Houston, TX 77030, USA
- Correspondence: ; Tel.: +1-713-792-2830
| | - Michael B. Atkins
- Department of Oncology, School of Medicine, Georgetown University, Washington, DC 20007, USA;
- Georgetown Lombardi Comprehensive Cancer Center, Washington, DC 20057, USA
| | - Simon Chowdhury
- Department of Medical Oncology, Guy’s and St Thomas’ Hospitals, London SE1 9RT, UK;
- Sarah Cannon Research Institute, London W1G 6AD, UK
| | - Paul Mainwaring
- Centre for Personalised Nanomedicine, The University of Queensland, Brisbane, QLD 4072, Australia;
| |
Collapse
|
82
|
Kim IH, Lee HJ. The Frontline Immunotherapy-Based Treatment of Advanced Clear Cell Renal Cell Carcinoma: Current Evidence and Clinical Perspective. Biomedicines 2022; 10:251. [PMID: 35203461 PMCID: PMC8869224 DOI: 10.3390/biomedicines10020251] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Accepted: 01/20/2022] [Indexed: 12/24/2022] Open
Abstract
Approximately 400,000 patients are diagnosed with kidney cancer annually worldwide, leading to approximately 170,000 deaths. Renal cell carcinoma (RCC) accounts for more than 90% of kidney cancers. The most common histological subtype is clear cell RCC, which is found in approximately 85% of metastatic RCC cases. The VHL-HIF-VEGF axis is well known; therefore, targeting VEGF has been the mainstay for managing advanced clear cell RCC. Recently, the treatment landscape for advanced clear cell RCC has changed extensively. In particular, immune checkpoint inhibitor-based treatment showed promising results in front-line treatment and became the standard of care. Herein, we review the current evidence on front-line treatment options and discuss the clinical and future perspective.
Collapse
Affiliation(s)
- In-Ho Kim
- Department of Internal Medicine, Division of Medical Oncology, Seoul St. Mary’s Hospital, The Catholic University of Korea, College of Medicine, Seoul 06591, Korea;
| | - Hyo Jin Lee
- Department of Internal Medicine, Chungnam National University School of Medicine, Daejeon 35015, Korea
| |
Collapse
|
83
|
Abstract
Actin is a highly conserved protein in mammals. The actin dynamics is regulated by actin-binding proteins and actin-related proteins. Nuclear actin and these regulatory proteins participate in multiple nuclear processes, including chromosome architecture organization, chromatin remodeling, transcription machinery regulation, and DNA repair. It is well known that the dysfunctions of these processes contribute to the development of cancer. Moreover, emerging evidence has shown that the deregulated actin dynamics is also related to cancer. This chapter discusses how the deregulation of nuclear actin dynamics contributes to tumorigenesis via such various nuclear events.
Collapse
Affiliation(s)
- Yuanjian Huang
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Shengzhe Zhang
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jae-Il Park
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center and Health Science Center, Houston, TX, USA.
- Program in Genetics and Epigenetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
84
|
Sobottka B, Nienhold R, Nowak M, Hench J, Haeuptle P, Frank A, Sachs M, Kahraman A, Moch H, Koelzer VH, Mertz KD. Integrated Analysis Of Immunotherapy Treated Clear Cell Renal Cell Carcinomas: An Exploratory Study. J Immunother 2022; 45:35-42. [PMID: 34406159 PMCID: PMC8654255 DOI: 10.1097/cji.0000000000000387] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 07/15/2021] [Indexed: 11/25/2022]
Abstract
Molecular or immunological differences between responders and nonresponders to immune checkpoint inhibitors (ICIs) of clear cell renal cell carcinomas (ccRCCs) remain incompletely understood. To address this question, we performed next-generation sequencing, methylation analysis, genome wide copy number analysis, targeted RNA sequencing and T-cell receptor sequencing, and we studied frequencies of tumor-infiltrating CD8+ T cells, presence of tertiary lymphoid structures (TLS) and PD-L1 expression in 8 treatment-naive ccRCC patients subsequently treated with ICI (3 responders, 5 nonresponders). Unexpectedly, we identified decreased frequencies of CD8+ tumor-infiltrating T cells and TLS, and a decreased expression of PD-L1 in ICI responders when compared with nonresponders. However, neither tumor-specific genetic alterations nor gene expression profiles correlated with response to ICI or the observed immune features. Our results underline the challenge to stratify ccRCC patients for immunotherapy based on routinely available pathologic primary tumor material, even with advanced technologies. Our findings emphasize the analysis of pretreated metastatic tissue in line with recent observations describing treatment effects on the tumor microenvironment. In addition, our data call for further investigation of additional parameters in a larger ccRCC cohort to understand the mechanistic implications of the observed differences in tumor-infiltrating CD8+ T cells, TLS, and PD-L1 expression.
Collapse
Affiliation(s)
- Bettina Sobottka
- Department of Pathology and Molecular Pathology, University Hospital Zurich, University of Zurich, Zurich
| | | | - Marta Nowak
- Department of Pathology and Molecular Pathology, University Hospital Zurich, University of Zurich, Zurich
| | | | | | | | | | - Abdullah Kahraman
- Department of Pathology and Molecular Pathology, University Hospital Zurich, University of Zurich, Zurich
| | - Holger Moch
- Department of Pathology and Molecular Pathology, University Hospital Zurich, University of Zurich, Zurich
| | - Viktor H. Koelzer
- Department of Pathology and Molecular Pathology, University Hospital Zurich, University of Zurich, Zurich
| | | |
Collapse
|
85
|
Kim TJ, Lee YH, Koo KC. Current and future perspectives on CAR-T cell therapy for renal cell carcinoma: A comprehensive review. Investig Clin Urol 2022; 63:486-498. [PMID: 36067994 PMCID: PMC9448669 DOI: 10.4111/icu.20220103] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 05/22/2022] [Accepted: 07/06/2022] [Indexed: 01/02/2023] Open
Abstract
In the clinical setting of renal cell carcinoma (RCC), immune reactions such as tumor-specific T cell responses can be spontaneous events or can be elicited by checkpoint inhibitors, cytokines, and other immunotherapy modalities. The results from immunotherapy have led to significant advances in treatment methods and patient outcomes. The approval of nivolumab primarily as a second-line monotherapy and the latest approval of novel combination therapies as first-line treatment have established the significance of immunotherapy in the treatment of RCC. In this perspective, chimeric antigen receptor (CAR)-T cell therapy represents a major advance in the developing field of immunotherapy. This treatment modality facilitates T cells to express specific CARs on the cell surface which are reinfused to the patient to treat the analogous tumor cells. After showing treatment potential in hematological malignancies, this new therapeutic approach has become a strong candidate as a therapeutic modality for solid neoplasms. Although CAR-T cell therapy has shown promise and clinical benefit compared to previous T-cell modulated immunotherapies, further studies are warranted to overcome unfavorable physiological settings and hindrances such as the lack of specific molecular targets, depletion of CAR-T cells, a hostile tumor microenvironment, and on/off-tumor toxicities. Several approaches are being considered and research is ongoing to overcome these problems. In this comprehensive review, we provide the rationale and preliminary results of CAR-T cell therapy in RCC and discuss emerging novel strategies and future directions.
Collapse
Affiliation(s)
- Tae Jin Kim
- Department of Urology, CHA University College of Medicine, CHA Bundang Medical Center, Seongnam, Korea
| | - Young Hwa Lee
- Department of Urology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Kyo Chul Koo
- Department of Urology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
86
|
Gantzer J, Davidson G, Vokshi B, Weingertner N, Bougoüin A, Moreira M, Lindner V, Lacroix G, Mascaux C, Chenard MP, Bertucci F, Davidson I, Kurtz JE, Sautès-Fridman C, Fridman WH, Malouf GG. OUP accepted manuscript. Oncologist 2022; 27:501-511. [PMID: 35278076 PMCID: PMC9177113 DOI: 10.1093/oncolo/oyac040] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 01/14/2022] [Indexed: 11/14/2022] Open
Abstract
Background Thoracic SMARCA4-deficient undifferentiated tumors (SMARCA4-UT) are aggressive neoplasms. Data linking BAF alterations with tumor microenvironment (TME) and efficacy of immune checkpoint inhibitors (ICI) are contradictory. The TME of SMARCA4-UT and their response to ICI are unknown. Materials and Methods Patients diagnosed with SMARCA4-UT in our institution were included. Immunostainings for tertiary lymphoid structures (TLS), immune cell markers, and checkpoints were assessed. Validation was performed using an independent transcriptome dataset including SMARCA4-UT, non–small cell lung cancers (NSCLC) with/without SMARCA4 mutations, and unclassified thoracic sarcomas (UTS). CXCL9 and PD-L1 expressions were assessed in NSCLC and thoracic fibroblast cell lines, with/without SMARCA4 knockdown, treated with/without interferon gamma. Results Nine patients were identified. All samples but one showed no TLS, consistent with an immune desert TME phenotype. Four patients received ICI as part of their treatment, but the only one who responded, had a tumor with a TLS and immune-rich TME. Unsupervised clustering of the validation cohort using immune cell scores identified 2 clusters associated with cell ontogeny and immunity (cluster 1 enriched for NSCLC independently of SMARCA4 status (n = 9/10; P = .001); cluster 2 enriched for SMARCA4-UT (n = 11/12; P = .005) and UTS (n = 5/5; P = .0005). SMARCA4 loss-of-function experiments revealed interferon-induced upregulation of CXCL9 and PD-L1 expression in the NSCLC cell line with no effect on the thoracic fibroblast cell line. Conclusion SMARCA4-UT mainly have an immune desert TME with limited efficacy to ICI. TME of SMARCA4-driven tumors varies according to the cell of origin questioning the interplay between BAF alterations, cell ontogeny and immunity.
Collapse
Affiliation(s)
- Justine Gantzer
- Corresponding author: Justine Gantzer, Department of Medical Oncology, Strasbourg-Europe Cancer Institute (ICANS), 17 rue Albert Calmette, 67033 Strasbourg, France. Tel: +33 3 68 76 72 25;
| | - Guillaume Davidson
- Department of Cancer and Functional Genomics, INSERM UMR_S1258, Institute of Genetics and of Molecular and Cellular Biology, Illkirch, France
| | - Bujamin Vokshi
- Department of Cancer and Functional Genomics, INSERM UMR_S1258, Institute of Genetics and of Molecular and Cellular Biology, Illkirch, France
| | - Noëlle Weingertner
- Fédération de Médecine Translationnelle (FMTS), Strasbourg, France
- Department of Pathology, University Hospital, Strasbourg, France
| | - Antoine Bougoüin
- Centre de recherche des Cordeliers, INSERM, Université de Paris, Sorbonne Université, Team 13- Complement, Inflammation and Cancer, Équipe labellisée Ligue contre le cancer, Paris, France
| | - Marco Moreira
- Centre de recherche des Cordeliers, INSERM, Université de Paris, Sorbonne Université, Team 13- Complement, Inflammation and Cancer, Équipe labellisée Ligue contre le cancer, Paris, France
| | - Véronique Lindner
- Fédération de Médecine Translationnelle (FMTS), Strasbourg, France
- Department of Pathology, University Hospital, Strasbourg, France
| | - Guillaume Lacroix
- Centre de recherche des Cordeliers, INSERM, Université de Paris, Sorbonne Université, Team 13- Complement, Inflammation and Cancer, Équipe labellisée Ligue contre le cancer, Paris, France
| | - Céline Mascaux
- Department of Pneumology, University Hospital, Strasbourg, France
- University of Strasbourg, Inserm UMR_S 1113, IRFAC, Laboratory Streinth (STress REsponse and INnovative THerapy against cancer), Strasbourg, France
| | - Marie-Pierre Chenard
- Fédération de Médecine Translationnelle (FMTS), Strasbourg, France
- Department of Pathology, University Hospital, Strasbourg, France
| | - François Bertucci
- Department of Medical Oncology, Cancer Research Center of Marseille (CRCM), INSERM U1068, CNRS UMR7258, Institut Paoli Calmettes, Aix-Marseille University, Marseille, France
| | - Irwin Davidson
- Department of Cancer and Functional Genomics, INSERM UMR_S1258, Institute of Genetics and of Molecular and Cellular Biology, Illkirch, France
| | - Jean-Emmanuel Kurtz
- Department of Medical Oncology, Strasbourg-Europe Cancer Institute (ICANS), Strasbourg, France
- Fédération de Médecine Translationnelle (FMTS), Strasbourg, France
| | - Catherine Sautès-Fridman
- Centre de recherche des Cordeliers, INSERM, Université de Paris, Sorbonne Université, Team 13- Complement, Inflammation and Cancer, Équipe labellisée Ligue contre le cancer, Paris, France
| | | | - Gabriel G Malouf
- Gabriel G. Malouf, Department of Medical Oncology, Strasbourg-Europe Cancer Institute (ICANS), 17 rue Albert Calmette, 67033 Strasbourg, France. Tel: +33 3 68 76 72 17;
| |
Collapse
|
87
|
Szarkowska J, Cwiek P, Szymanski M, Rusetska N, Jancewicz I, Stachowiak M, Swiatek M, Luba M, Konopinski R, Kubala S, Zub R, Kucharz J, Wiechno P, Siedlecki JA, Markowicz S, Sarnowska E, Sarnowski TJ. RRM2 gene expression depends on BAF180 subunit of SWISNF chromatin remodeling complex and correlates with abundance of tumor infiltrating lymphocytes in ccRCC. Am J Cancer Res 2021; 11:5965-5978. [PMID: 35018236 PMCID: PMC8727810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Accepted: 10/12/2021] [Indexed: 06/14/2023] Open
Abstract
About 40% of clear cell renal cell carcinoma (ccRCC) cases carry the pbrm1 mutation inactivating BAF180 subunit of the SWI/SNF chromatin remodeling complex (CRC). Here we show that the majority of transcriptomic changes appear at the stage I of ccRCC development. By contrast, the stage II ccRCC exhibits hyperactivation of DNA replication demonstrated by the overexpression of several genes, e.g., RRM1 and RRM2 genes encoding subunits of ribonucleotide reductase (RNR) complex. We found that the degree of RRM1 and RRM2 upregulation in ccRCC patients depends on pbrm1 mutation. We show that the BAF180 protein product of the PBRM1 gene directly binds to RRM1 and RRM2 loci. The BAF180 binding regions are targeted by regulatory proteins previously reported as SWI/SNF CRC interacting partners. BAF180 binding to RRMs loci correlates with enrichment of H3K27me3 in case of RRM1 and H3K14Ac on RRM2, indicating the existence of differential regulatory mechanism controlling expression of these genes. We found that the strong overexpression of RRM2 in ccRCC patient samples correlates with T cell infiltration. Surprisingly, the majority of tumor infiltrating lymphocytes (TILs) consisted of CD4+ T cells. Furthermore, we show that exhausted CD4+ T cells induced the expression of the RRM2 gene in the primary ccRCC cell line. Collectively, our results provide the link between PBRM1 loss, RRM2 expression and T cell infiltration, which may lead to the establishment of new treatment of this disease.
Collapse
Affiliation(s)
- Joanna Szarkowska
- Department of Experimental Immunotherapy, Maria Sklodowska-Curie National Research Institute of OncologyWarsaw, Poland
| | - Pawel Cwiek
- Institute of Biochemistry and Biophysics, Polish Academy of SciencesWarsaw, Poland
| | - Michal Szymanski
- Department of Urology and Urological Oncology, Central Clinical Hospital of Ministry of the Interior and Administration in WarsawWarsaw, Poland
| | - Natalia Rusetska
- Department of Experimental Immunotherapy, Maria Sklodowska-Curie National Research Institute of OncologyWarsaw, Poland
| | - Iga Jancewicz
- Department of Experimental Immunotherapy, Maria Sklodowska-Curie National Research Institute of OncologyWarsaw, Poland
| | - Malgorzata Stachowiak
- Department of Experimental Immunotherapy, Maria Sklodowska-Curie National Research Institute of OncologyWarsaw, Poland
| | - Monika Swiatek
- Department of Experimental Immunotherapy, Maria Sklodowska-Curie National Research Institute of OncologyWarsaw, Poland
| | - Maciej Luba
- Department of Experimental Immunotherapy, Maria Sklodowska-Curie National Research Institute of OncologyWarsaw, Poland
| | - Ryszard Konopinski
- Department of Experimental Immunotherapy, Maria Sklodowska-Curie National Research Institute of OncologyWarsaw, Poland
| | - Szymon Kubala
- Institute of Biochemistry and Biophysics, Polish Academy of SciencesWarsaw, Poland
| | - Renata Zub
- Department of Experimental Immunotherapy, Maria Sklodowska-Curie National Research Institute of OncologyWarsaw, Poland
| | - Jakub Kucharz
- Department of Uro-oncology, Maria Sklodowska-Curie National Research Institute of OncologyWarsaw, Poland
| | - Pawel Wiechno
- Department of Uro-oncology, Maria Sklodowska-Curie National Research Institute of OncologyWarsaw, Poland
| | - Janusz A Siedlecki
- Department of Molecular and Translational Oncology, Maria Sklodowska-Curie National Research Institute of OncologyWarsaw, Poland
| | - Sergiusz Markowicz
- Department of Experimental Immunotherapy, Maria Sklodowska-Curie National Research Institute of OncologyWarsaw, Poland
| | - Elzbieta Sarnowska
- Department of Experimental Immunotherapy, Maria Sklodowska-Curie National Research Institute of OncologyWarsaw, Poland
| | - Tomasz J Sarnowski
- Institute of Biochemistry and Biophysics, Polish Academy of SciencesWarsaw, Poland
| |
Collapse
|
88
|
Kim EJ, Liu P, Zhang S, Donahue K, Wang Y, Schehr J, Wolfe S, Dickerson A, Lu L, Rui L, Zhong X, Wisinski K, Yu M, Suzuki A, Lang J, Ong I, Xu W. BAF155 methylation drives metastasis by hijacking super-enhancers and subverting anti-tumor immunity. Nucleic Acids Res 2021; 49:12211-12233. [PMID: 34865122 PMCID: PMC8643633 DOI: 10.1093/nar/gkab1122] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 10/06/2021] [Accepted: 10/26/2021] [Indexed: 11/25/2022] Open
Abstract
Subunits of the chromatin remodeler SWI/SNF are the most frequently disrupted genes in cancer. However, how post-translational modifications (PTM) of SWI/SNF subunits elicit epigenetic dysfunction remains unknown. Arginine-methylation of BAF155 by coactivator-associated arginine methyltransferase 1 (CARM1) promotes triple-negative breast cancer (TNBC) metastasis. Herein, we discovered the dual roles of methylated-BAF155 (me-BAF155) in promoting tumor metastasis: activation of super-enhancer-addicted oncogenes by recruiting BRD4, and repression of interferon α/γ pathway genes to suppress host immune response. Pharmacological inhibition of CARM1 and BAF155 methylation not only abrogated the expression of an array of oncogenes, but also boosted host immune responses by enhancing the activity and tumor infiltration of cytotoxic T cells. Moreover, strong me-BAF155 staining was detected in circulating tumor cells from metastatic cancer patients. Despite low cytotoxicity, CARM1 inhibitors strongly inhibited TNBC cell migration in vitro, and growth and metastasis in vivo. These findings illustrate a unique mechanism of arginine methylation of a SWI/SNF subunit that drives epigenetic dysregulation, and establishes me-BAF155 as a therapeutic target to enhance immunotherapy efficacy.
Collapse
Affiliation(s)
- Eui-Jun Kim
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI 53706, USA
- Carbone Comprehensive Cancer Center, University of Wisconsin, Madison, WI 53706, USA
| | - Peng Liu
- Department of Biostatistics and Medical Informatics. School of Medicine and Public Health, University of Wisconsin, Madison, WI 53706, USA
- Carbone Comprehensive Cancer Center, University of Wisconsin, Madison, WI 53706, USA
| | - Shengjie Zhang
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Kristine Donahue
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI 53706, USA
- Carbone Comprehensive Cancer Center, University of Wisconsin, Madison, WI 53706, USA
| | - Yidan Wang
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI 53706, USA
- Carbone Comprehensive Cancer Center, University of Wisconsin, Madison, WI 53706, USA
| | - Jennifer L Schehr
- Department of Medicine, University of Wisconsin, Madison, WI 53706, USA
- Carbone Comprehensive Cancer Center, University of Wisconsin, Madison, WI 53706, USA
| | - Serena K Wolfe
- Department of Medicine, University of Wisconsin, Madison, WI 53706, USA
- Carbone Comprehensive Cancer Center, University of Wisconsin, Madison, WI 53706, USA
| | - Amber Dickerson
- Department of Stem Cell Biology and Regenerative Medicine, and USC Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Li Lu
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI 53715, USA
- Laboratory of Genetics, University of Wisconsin-Madison, Madison WI, USA
| | - Lixin Rui
- Department of Obstetrics and Gynecology, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53706, USA
- Carbone Comprehensive Cancer Center, University of Wisconsin, Madison, WI 53706, USA
| | - Xuehua Zhong
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI 53715, USA
- Laboratory of Genetics, University of Wisconsin-Madison, Madison WI, USA
| | - Kari B Wisinski
- Department of Medicine, University of Wisconsin, Madison, WI 53706, USA
- Carbone Comprehensive Cancer Center, University of Wisconsin, Madison, WI 53706, USA
| | - Min Yu
- Department of Stem Cell Biology and Regenerative Medicine, and USC Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Aussie Suzuki
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI 53706, USA
- Carbone Comprehensive Cancer Center, University of Wisconsin, Madison, WI 53706, USA
| | - Joshua M Lang
- Department of Medicine, University of Wisconsin, Madison, WI 53706, USA
- Carbone Comprehensive Cancer Center, University of Wisconsin, Madison, WI 53706, USA
| | - Irene M Ong
- Department of Obstetrics and Gynecology, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53706, USA
- Department of Biostatistics and Medical Informatics. School of Medicine and Public Health, University of Wisconsin, Madison, WI 53706, USA
- Carbone Comprehensive Cancer Center, University of Wisconsin, Madison, WI 53706, USA
| | - Wei Xu
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI 53706, USA
- Carbone Comprehensive Cancer Center, University of Wisconsin, Madison, WI 53706, USA
| |
Collapse
|
89
|
Fountzilas E, Kurzrock R, Vo HH, Tsimberidou AM. Wedding of Molecular Alterations and Immune Checkpoint Blockade: Genomics as a Matchmaker. J Natl Cancer Inst 2021; 113:1634-1647. [PMID: 33823006 PMCID: PMC9890928 DOI: 10.1093/jnci/djab067] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/21/2021] [Accepted: 03/10/2021] [Indexed: 02/05/2023] Open
Abstract
The development of checkpoint blockade immunotherapy has transformed the medical oncology armamentarium. But despite its favorable impact on clinical outcomes, immunotherapy benefits only a subset of patients, and a substantial proportion of these individuals eventually manifest resistance. Serious immune-related adverse events and hyperprogression have also been reported. It is therefore essential to understand the molecular mechanisms and identify the drivers of therapeutic response and resistance. In this review, we provide an overview of the current and emerging clinically relevant genomic biomarkers implicated in checkpoint blockade outcome. US Food and Drug Administration-approved molecular biomarkers of immunotherapy response include mismatch repair deficiency and/or microsatelliteinstability and tumor mutational burden of at least 10 mutations/megabase. Investigational genomic-associated biomarkers for immunotherapy response include alterations of the following genes/associated pathways: chromatin remodeling (ARID1A, PBRM1, SMARCA4, SMARCB1, BAP1), major histocompatibility complex, specific (eg, ultraviolet, APOBEC) mutational signatures, T-cell receptor repertoire, PDL1, POLE/POLD1, and neo-antigens produced by the mutanome, those potentially associated with resistance include β2-microglobulin, EGFR, Keap1, JAK1/JAK2/interferon-gamma signaling, MDM2, PTEN, STK11, and Wnt/Beta-catenin pathway alterations. Prospective clinical trials are needed to assess the role of a composite of these biomarkers to optimize the implementation of precision immunotherapy in patient care.
Collapse
Affiliation(s)
- Elena Fountzilas
- Department of Medical Oncology, Euromedica General Clinic, Thessaloniki, Greece
- European University Cyprus, Limassol, Cyprus
| | - Razelle Kurzrock
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, UC San Diego Moores Cancer Center, San Diego, CA, USA
| | - Henry Hiep Vo
- The University of Texas MD Anderson Cancer Center, Department of Investigational Cancer Therapeutics, Houston, TX, USA
| | - Apostolia-Maria Tsimberidou
- The University of Texas MD Anderson Cancer Center, Department of Investigational Cancer Therapeutics, Houston, TX, USA
| |
Collapse
|
90
|
Anker J, Miller J, Taylor N, Kyprianou N, Tsao CK. From Bench to Bedside: How the Tumor Microenvironment Is Impacting the Future of Immunotherapy for Renal Cell Carcinoma. Cells 2021; 10:3231. [PMID: 34831452 PMCID: PMC8619121 DOI: 10.3390/cells10113231] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 11/16/2021] [Accepted: 11/17/2021] [Indexed: 12/23/2022] Open
Abstract
Immunotherapy has revolutionized the treatment landscape for many cancer types. The treatment for renal cell carcinoma (RCC) has especially evolved in recent years, from cytokine-based immunotherapies to immune checkpoint inhibitors. Although clinical benefit from immunotherapy is limited to a subset of patients, many combination-based approaches have led to improved outcomes. The success of such approaches is a direct result of the tumor immunology knowledge accrued regarding the RCC microenvironment, which, while highly immunogenic, demonstrates many unique characteristics. Ongoing translational work has elucidated some of the mechanisms of response, as well as primary and secondary resistance, to immunotherapy. Here, we provide a comprehensive review of the RCC immunophenotype with a specific focus on how preclinical and clinical data are shaping the future of immunotherapy.
Collapse
Affiliation(s)
- Jonathan Anker
- Division of Internal Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
| | - Justin Miller
- Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (J.M.); (N.T.)
| | - Nicole Taylor
- Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (J.M.); (N.T.)
| | - Natasha Kyprianou
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
- Department of Pathology and Molecular and Cell Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Che-Kai Tsao
- Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (J.M.); (N.T.)
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
| |
Collapse
|
91
|
Aftimos P, Oliveira M, Irrthum A, Fumagalli D, Sotiriou C, Gal-Yam EN, Robson ME, Ndozeng J, Di Leo A, Ciruelos EM, de Azambuja E, Viale G, Scheepers ED, Curigliano G, Bliss JM, Reis-Filho JS, Colleoni M, Balic M, Cardoso F, Albanell J, Duhem C, Marreaud S, Romagnoli D, Rojas B, Gombos A, Wildiers H, Guerrero-Zotano A, Hall P, Bonetti A, Larsson KF, Degiorgis M, Khodaverdi S, Greil R, Sverrisdóttir Á, Paoli M, Seyll E, Loibl S, Linderholm B, Zoppoli G, Davidson NE, Johannsson OT, Bedard PL, Loi S, Knox S, Cameron DA, Harbeck N, Montoya ML, Brandão M, Vingiani A, Caballero C, Hilbers FS, Yates LR, Benelli M, Venet D, Piccart MJ. Genomic and Transcriptomic Analyses of Breast Cancer Primaries and Matched Metastases in AURORA, the Breast International Group (BIG) Molecular Screening Initiative. Cancer Discov 2021; 11:2796-2811. [PMID: 34183353 PMCID: PMC9414283 DOI: 10.1158/2159-8290.cd-20-1647] [Citation(s) in RCA: 116] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 04/05/2021] [Accepted: 06/11/2021] [Indexed: 02/01/2023]
Abstract
AURORA aims to study the processes of relapse in metastatic breast cancer (MBC) by performing multi-omics profiling on paired primary tumors and early-course metastases. Among 381 patients (primary tumor and metastasis pairs: 252 targeted gene sequencing, 152 RNA sequencing, 67 single nucleotide polymorphism arrays), we found a driver role for GATA1 and MEN1 somatic mutations. Metastases were enriched in ESR1, PTEN, CDH1, PIK3CA, and RB1 mutations; MDM4 and MYC amplifications; and ARID1A deletions. An increase in clonality was observed in driver genes such as ERBB2 and RB1. Intrinsic subtype switching occurred in 36% of cases. Luminal A/B to HER2-enriched switching was associated with TP53 and/or PIK3CA mutations. Metastases had lower immune score and increased immune-permissive cells. High tumor mutational burden correlated to shorter time to relapse in HR+/HER2- cancers. ESCAT tier I/II alterations were detected in 51% of patients and matched therapy was used in 7%. Integration of multi-omics analyses in clinical practice could affect treatment strategies in MBC. SIGNIFICANCE: The AURORA program, through the genomic and transcriptomic analyses of matched primary and metastatic samples from 381 patients with breast cancer, coupled with prospectively collected clinical data, identified genomic alterations enriched in metastases and prognostic biomarkers. ESCAT tier I/II alterations were detected in more than half of the patients.This article is highlighted in the In This Issue feature, p. 2659.
Collapse
Affiliation(s)
- Philippe Aftimos
- Institut Jules Bordet - Université Libre de Bruxelles, Brussels, Belgium
| | - Mafalda Oliveira
- Medical Oncology Department, Vall d'Hebron University Hospital, Barcelona, Spain
| | | | | | - Christos Sotiriou
- Institut Jules Bordet - Université Libre de Bruxelles, Brussels, Belgium
| | | | - Mark E Robson
- Memorial Sloan Kettering Cancer Center, New York, New York
| | - Justin Ndozeng
- Institut Jules Bordet - Université Libre de Bruxelles, Brussels, Belgium
| | | | | | | | - Giuseppe Viale
- IEO, Istituto Europeo di Oncologia, IRCCS, and University of Milan, Milan, Italy
| | | | - Giuseppe Curigliano
- IEO, Istituto Europeo di Oncologia, IRCCS, and University of Milan, Milan, Italy
| | - Judith M Bliss
- The Institute of Cancer Research, London, United Kingdom
| | | | - Marco Colleoni
- IEO, Istituto Europeo di Oncologia, IRCCS, and University of Milan, Milan, Italy
| | | | - Fatima Cardoso
- Breast Unit, Champalimaud Clinical Center/Champalimaud Foundation, Lisbon, Portugal
| | - Joan Albanell
- Hospital del Mar - CIBERONC; IMIM, Barcelona; Pompeu Fabra University, Barcelona, Spain
| | - Caroline Duhem
- Centre Hospitalier Luxembourg, Luxembourg City, Luxembourg
| | | | | | - Beatriz Rojas
- CIOCC (Centro Integral Oncologico "Clara Campal"), Madrid, Spain
| | - Andrea Gombos
- Institut Jules Bordet - Université Libre de Bruxelles, Brussels, Belgium
| | | | | | - Peter Hall
- University of Edinburgh Cancer Research Centre, Edinburgh, United Kingdom
| | - Andrea Bonetti
- Department of Oncology AZIENDA ULSS 9 Verona, Verona, Italy
| | | | | | - Silvia Khodaverdi
- Sana Klinikum Offenbach, Klinik für Gynaekologie und Geburtshilfe, Offenbach, Germany
| | - Richard Greil
- Paracelsus Medical University Salzburg, Salzburg Cancer Research Institute-CCCIT and Cancer Cluster Salzburg, Salzburg, Austria
| | | | | | - Ethel Seyll
- Institut Jules Bordet - Université Libre de Bruxelles, Brussels, Belgium
| | | | | | - Gabriele Zoppoli
- Università degli Studi di Genova and IRCCS Ospedale Policlinico San Martino, San Martino, Italy
| | - Nancy E Davidson
- Fred Hutchinson Cancer Research Center and University of Washington, Seattle, Washington
| | | | | | - Sherene Loi
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Susan Knox
- Europa Donna- The European Breast Cancer Coalition, Milan, Italy
| | - David A Cameron
- University of Edinburgh Cancer Research Centre, Edinburgh, United Kingdom
| | - Nadia Harbeck
- Breast Center, LMU University Hospital, Munich, Germany, and West German Study Group, Moenchengladbach, Germany
| | | | - Mariana Brandão
- Institut Jules Bordet - Université Libre de Bruxelles, Brussels, Belgium
| | - Andrea Vingiani
- Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | | | | | - Lucy R Yates
- Wellcome Trust Sanger Institute, London, United Kingdom
| | | | - David Venet
- Institut Jules Bordet - Université Libre de Bruxelles, Brussels, Belgium
| | - Martine J Piccart
- Institut Jules Bordet - Université Libre de Bruxelles, Brussels, Belgium.
- Breast International Group, Brussels, Belgium
| |
Collapse
|
92
|
Chabanon RM, Rouanne M, Lord CJ, Soria JC, Pasero P, Postel-Vinay S. Targeting the DNA damage response in immuno-oncology: developments and opportunities. Nat Rev Cancer 2021; 21:701-717. [PMID: 34376827 DOI: 10.1038/s41568-021-00386-6] [Citation(s) in RCA: 167] [Impact Index Per Article: 41.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/22/2021] [Indexed: 02/07/2023]
Abstract
Immunotherapy has revolutionized cancer treatment and substantially improved patient outcome with regard to multiple tumour types. However, most patients still do not benefit from such therapies, notably because of the absence of pre-existing T cell infiltration. DNA damage response (DDR) deficiency has recently emerged as an important determinant of tumour immunogenicity. A growing body of evidence now supports the concept that DDR-targeted therapies can increase the antitumour immune response by (1) promoting antigenicity through increased mutability and genomic instability, (2) enhancing adjuvanticity through the activation of cytosolic immunity and immunogenic cell death and (3) favouring reactogenicity through the modulation of factors that control the tumour-immune cell synapse. In this Review, we discuss the interplay between the DDR and anticancer immunity and highlight how this dynamic interaction contributes to shaping tumour immunogenicity. We also review the most innovative preclinical approaches that could be used to investigate such effects, including recently developed ex vivo systems. Finally, we highlight the therapeutic opportunities presented by the exploitation of the DDR-anticancer immunity interplay, with a focus on those in early-phase clinical development.
Collapse
Affiliation(s)
- Roman M Chabanon
- ATIP-Avenir Group, Inserm Unit U981, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France
- The CRUK Gene Function Laboratory and Breast Cancer Now Toby Robins Breast Cancer Research Centre, The Institute of Cancer Research, London, UK
| | - Mathieu Rouanne
- Equipe Labellisée Ligue Nationale contre le Cancer, Inserm Unit U1015, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France
- Département d'Urologie, Hôpital Foch, Université Versailles-Saint-Quentin-en-Yvelines, Université Paris-Saclay, Suresnes, France
| | - Christopher J Lord
- The CRUK Gene Function Laboratory and Breast Cancer Now Toby Robins Breast Cancer Research Centre, The Institute of Cancer Research, London, UK
| | - Jean-Charles Soria
- Drug Development Department (DITEP), Gustave Roussy Cancer Campus, Villejuif, France
- Faculté de Médicine, Université Paris-Sud, Université Paris-Saclay, Le Kremlin Bicêtre, France
| | - Philippe Pasero
- Equipe Labellisée Ligue contre le Cancer, Institut de Génétique Humaine, CNRS, Université de Montpellier, Montpellier, France
| | - Sophie Postel-Vinay
- ATIP-Avenir Group, Inserm Unit U981, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France.
- Drug Development Department (DITEP), Gustave Roussy Cancer Campus, Villejuif, France.
- Faculté de Médicine, Université Paris-Sud, Université Paris-Saclay, Le Kremlin Bicêtre, France.
| |
Collapse
|
93
|
Botta GP, Kato S, Patel H, Fanta P, Lee S, Okamura R, Kurzrock R. SWI/SNF complex alterations as a biomarker of immunotherapy efficacy in pancreatic cancer. JCI Insight 2021; 6:e150453. [PMID: 34375311 PMCID: PMC8492298 DOI: 10.1172/jci.insight.150453] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 08/04/2021] [Indexed: 12/23/2022] Open
Abstract
BACKGROUNDImmune checkpoint inhibitors (ICIs) fail to demonstrate efficacy in pancreatic cancer. Recently, genomic biomarkers have been associated with response to ICIs: microsatellite instability high (MSI-H) and tumor mutation burden (TMB) > 10 mutations/Mb. Alterations in Switch/Sucrose Nonfermentable (SWI/SNF) chromatin remodeling genes may predispose to improved outcomes with immunotherapy. The current study examined a possible role for SWI/SNF complex abnormalities in pancreatic cancer responsiveness to ICIs.METHODSA database of 6831 cancer patients that had undergone next-generation sequencing (NGS) was filtered for advanced pancreatic cancer, SWI/SNF alterations, and outcomes depending on immunotherapy treatment.RESULTSNine patients had metastatic pancreatic adenocarcinoma harboring SWI/SNF chromatin remodeling gene alterations and had received ICIs: 7 had an ARID1A alteration (77%); 2, ARID1B (22%); 3, SMARCA4 (33%); 1, SMARCB1 (11%); and 1, PBRM1 (11%). Three patients possessed more than 1 SWI/SNF complex alteration. Only 3 tumors were microsatellite unstable. Eight of 9 patients (89%) achieved an objective response, including a complete remission, with the 2 longest responses ongoing at 33+ and 36+ months. Median progression-free and overall survival was 9 and 15 months, respectively. Responses occurred even in the presence of microsatellite stability, low TMB, and/or low PD-L1 expression.CONCLUSIONA small subset of patients with pancreatic cancer have genomic alterations in SWI/SNF chromatin remodeling components and appear to be responsive to ICIs, suggesting the need for prospective trials.TRIAL REGISTRATIONClinicalTrials.gov, NCT02478931.FUNDINGJoan and Irwin Jacobs Fund, NIH P30 CA023100 (RK) and LRP KYGF9753 (GPB), the Gershenson, Duarte, and anonymous patient families (GPB).
Collapse
Affiliation(s)
- Gregory P. Botta
- Center for Personalized Cancer Therapy, Department of Medicine, and
- Division of Hematology/Oncology, Department of Medicine, UCSD, La Jolla, California, USA
| | - Shumei Kato
- Center for Personalized Cancer Therapy, Department of Medicine, and
- Division of Hematology/Oncology, Department of Medicine, UCSD, La Jolla, California, USA
| | - Hitendra Patel
- Division of Hematology/Oncology, Department of Medicine, UCSD, La Jolla, California, USA
| | - Paul Fanta
- Division of Hematology/Oncology, Department of Medicine, UCSD, La Jolla, California, USA
| | - Suzanna Lee
- Center for Personalized Cancer Therapy, Department of Medicine, and
| | - Ryosuke Okamura
- Department of Surgery, Kyoto University Hospital, Kyoto, Japan
| | - Razelle Kurzrock
- Center for Personalized Cancer Therapy, Department of Medicine, and
- Division of Hematology/Oncology, Department of Medicine, UCSD, La Jolla, California, USA
| |
Collapse
|
94
|
Zeuschner P, Zaccagnino A, Junker K. [Biomarkers for renal cell tumours]. Aktuelle Urol 2021; 52:452-463. [PMID: 34157774 DOI: 10.1055/a-1517-6259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
During the last three decades, renal tumours have become increasingly well differentiated on the basis of their histopathological and molecular features. This subtyping has increasingly impacted clinical practice because more therapeutic options are available in organ-confined and metastatic renal cell tumours. The knowledge of the underlying molecular alterations is essential to develop molecular targeted therapies and to select the most effective systemic therapy for each patient. This manuscript gives an overview of the molecular differentiation on the one hand, and on diagnostic, prognostic and predictive biomarkers on the other hand.
Collapse
Affiliation(s)
- Philip Zeuschner
- Klinik für Urologie und Kinderurologie, Universitätsklinikum des Saarlandes, Homburg/Saar
| | - Angela Zaccagnino
- Klinik für Urologie und Kinderurologie, Universitätsklinikum des Saarlandes, Homburg/Saar
| | - Kerstin Junker
- Klinik für Urologie und Kinderurologie, Universitätsklinikum des Saarlandes, Homburg/Saar
| |
Collapse
|
95
|
Aili A, Wen J, Xue L, Wang J. Mutational Analysis of PBRM1 and Significance of PBRM1 Mutation in Anti-PD-1 Immunotherapy of Clear Cell Renal Cell Carcinoma. Front Oncol 2021; 11:712765. [PMID: 34447697 PMCID: PMC8383204 DOI: 10.3389/fonc.2021.712765] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 07/23/2021] [Indexed: 01/22/2023] Open
Abstract
Renal cell carcinoma is a common solid tumor. PBRM1 is one of the most mutation-prone genes in clear cell renal cell carcinoma (ccRCC) with the occurrence of mutation in 40% of ccRCC patients. Mutations in PBRM1 have been correlated with the efficacy of immunotherapy. However, the mutation types of PBRM1 are not well characterized. The effects of PBRM1 expression levels in the tumor microenvironment are not well studied. In addition, the mechanism and effect of anti-PD-1 immunotherapy in ccRCC tumor microenvironments are not well clarified. In this study, using bioinformatics methods we analyzed the alternation frequency and expression levels of PBRM1 in various tumors. Next, we experimentally validated their expression levels in ccRCC tissues from human and mouse models. We attempted to clarify the mechanisms of anti-PD-1 immunotherapy in ccRCC with various PBRM1 expression levels. Our results showed that deficiency of PBRM1 protein is correlated with CD4 T cell reduction in human and mouse ccRCC tissues. We also showed that anti-PD-1 Immunotherapy can increase the infiltration of T cells in both PBRM1 high and PBRM1 low tumors but to different degrees. Our study indicates that the reduction of CD4 cells in tumor tissues with low expression of PBRM1 may explain the compromised efficacy of anti-PD-1 immunotherapy in patients with PBRM1 mutated ccRCC. Our study sheds light on the potential of PBRM1 as a therapeutic target in ccRCC.
Collapse
Affiliation(s)
| | - Jie Wen
- Department of Radiation Oncology, Peking University Third Hospital, Beijing, China
| | - Lixiang Xue
- Department of Radiation Oncology, Peking University Third Hospital, Beijing, China.,Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China
| | - Junjie Wang
- Department of Radiation Oncology, Peking University Third Hospital, Beijing, China
| |
Collapse
|
96
|
Wei J, Lu J, Cao Y, Yao G, Huang Y, Zhao H, Pan Y, Feng Z, Chen Z, Chen W, Luo J, Cao J. DDX39B Predicts Poor Survival and Associated with Clinical Benefit of Anti-PD-L1 Therapy in ccRCC. Curr Cancer Drug Targets 2021; 21:849-859. [PMID: 34382524 DOI: 10.2174/1568009621666210811115054] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Revised: 04/08/2021] [Accepted: 04/25/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICI) have been shown to improve overall survival (OS) in clear cell renal cell carcinoma (ccRCC) patients. However, less than half of the ccRCC patients have objective response to ICI. OBJECTIVE We aim to assess the role of DDX39B in predicting ccRCC patients' OS and ICI therapy response. METHODS DDX39B was detected by immunohistochemistry in a tissue microarray of 305 ccRCC patients. DDX39B and its relationship with the prognosis of ccRCC were also evaluated in TCGA set and a RECA-EU set. The expression of DDX39B and patients survival was also analysed in two datasets of ccRCC patients treated with ICI. RESULTS Overexpression of DDX39B predicted poor OS of ccRCC patients in SYSU set, TCGA set, and a RECA-EU set. DDX39B expression was significantly positive with the expression of PD-L1 and other immunomodulators., DDX39B negatively correlated with cytotoxic T-lymphocyte and HDAC10 exon 3 inclusion in ccRCC. DDX39B knockdown decreased the expression of PD-L1 and increased the expression of HDAC10 exon 3 in renal cancer ACHN cells. Patients of ccRCC with lower levels of HDAC10 exon 3 inclusion have higher TNM stage, higher Fuhrman grade and poor OS. There was a tendency that patients with DDX39B high expression had longer OS and PFS than patients with DDX39B low expression in ccRCC patients treated with ICI. CONCLUSION DDX39B gene is highly expressed in ccRCC and is closely related to patients' OS. DDX39B might increase PD-L1 expression via the enhancement of HDAC10 exon 3 skipping, thereby promoting the ICI therapy response.
Collapse
Affiliation(s)
- Jinhuan Wei
- Department of Urology, First Affiliated Hospital, Sun Yat-sen University, Guangdong. China
| | - Jun Lu
- Department of Urology, First Affiliated Hospital, Sun Yat-sen University, Guangdong. China
| | - Yun Cao
- Department of Pathology, Cancer Center, Sun Yat-sen University, Guangzhou, Guangdong. China
| | - Gaosheng Yao
- Department of Urology, First Affiliated Hospital, Sun Yat-sen University, Guangdong. China
| | - Yong Huang
- Department of Urology, First Affiliated Hospital, Sun Yat-sen University, Guangdong. China
| | - Hongwei Zhao
- Department of Urology, Affiliated Yantai Yuhuangding Hospital, Qingdao University Medical College, Yantai. China
| | - Yihui Pan
- Department of Urology, First Affiliated Hospital, Sun Yat-sen University, Guangdong. China
| | - Zihao Feng
- Department of Urology, First Affiliated Hospital, Sun Yat-sen University, Guangdong. China
| | - Zhenhua Chen
- Department of Urology, First Affiliated Hospital, Sun Yat-sen University, Guangdong. China
| | - Wei Chen
- Department of Urology, First Affiliated Hospital, Sun Yat-sen University, Guangdong. China
| | - Junhang Luo
- Department of Urology, First Affiliated Hospital, Sun Yat-sen University, Guangdong. China
| | - Jiazheng Cao
- Department of Urology, Jiangmen Central Hospital, Affiliated Jiangmen Hospital of Sun Yet-sen University, Jiangmen, Guangdong. China
| |
Collapse
|
97
|
Hagiwara M, Fushimi A, Yamashita N, Bhattacharya A, Rajabi H, Long MD, Yasumizu Y, Oya M, Liu S, Kufe D. MUC1-C activates the PBAF chromatin remodeling complex in integrating redox balance with progression of human prostate cancer stem cells. Oncogene 2021; 40:4930-4940. [PMID: 34163028 PMCID: PMC8321896 DOI: 10.1038/s41388-021-01899-y] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 05/20/2021] [Accepted: 06/07/2021] [Indexed: 12/19/2022]
Abstract
The polybromo-associated PBAF (SWI/SNF) chromatin remodeling complex, which includes PBRM1, ARID2, and BRD7, regulates cell differentiation and genomic integrity. MUC1-C is an oncogenic protein that drives lineage plasticity in prostate cancer (PC) progression. The present work demonstrates that MUC1-C induces PBRM1, ARID2, and BRD7 expression by the previously unrecognized E2F1-mediated activation of their respective promoters. The functional significance of the MUC1-C→PBAF pathway is supported by demonstrating involvement of MUC1-C in associating with nuclear PBAF and driving the NRF2 antioxidant gene transcriptome in PC cells. Mechanistically, MUC1-C forms a complex with NRF2 and PBRM1 on the NRF2 target SLC7A11 gene that encodes the xCT cystine-glutamate antiporter, increases chromatin accessibility and induces SLC7A11/xCT expression. We also show that MUC1-C and PBRM1 are necessary for induction of other NRF2 target genes, including G6PD and PGD that regulate the pentose phosphate pathway. Our results further demonstrate that MUC1-C integrates activation of PBRM1 with the regulation of antioxidant genes, ROS levels, pluripotency factor expression and the cancer stem cell (CSC) state. These findings reveal a role for MUC1-C in regulating PBAF, redox balance and lineage plasticity of PC CSC progression. Our findings also uncover involvement of MUC1-C in integrating the PBAF and BAF pathways in cancer.
Collapse
Affiliation(s)
- Masayuki Hagiwara
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Atsushi Fushimi
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Nami Yamashita
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | | | - Hasan Rajabi
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Mark D Long
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Yota Yasumizu
- Department of Urology, Keio University School of Medicine, Tokyo, Japan
| | - Mototsugu Oya
- Department of Urology, Keio University School of Medicine, Tokyo, Japan
| | - Song Liu
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Donald Kufe
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
98
|
Dias Carneiro APC, Marques Monteiro FS, Soares A. PBRM1 Mutations as a Predictive Biomarker for Immunotherapy in Metastatic Renal Cell Carcinoma: A Systematic Review. KIDNEY CANCER 2021. [DOI: 10.3233/kca-210111] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
INTRODUCTION: Genomic features linked to prediction of response to immunotherapy in metastatic renal cell carcinoma (mRCC) are still lacking. Protein polybromo-1 (PBRM1) mutations have been studied as a potential biomarker of clinical benefit, with conflicting published data so far. MATERIAL AND METHODS: This systematic review was guided by the standards of the PRISMA statement to identify studies involving mRCC, immunotherapy and mutations in PBRM1. The main objective was to assess the relationship between PBRM1 mutations and response to immune checkpoint inhibitors (ICI) in patients with mRCC. RESULTS: After an initial search that identified 422 studies, 8 studies met the eligibility criteria and were selected for the final analysis. Data are included from 2 trials in the first-line treatment setting, and 6 trials in second- or later treatment lines evaluating the relationship between the presence of PBRM1 mutations and clinical benefit (CB) with ICI treatment. Regarding the first-line treatment setting, the analysis of both studies failed to show any CB in patients with PBRM1 mutations treated with ICI. However, for the second- and later treatment lines, the results were mixed. CONCLUSIONS: PBRM1 mutations may be a potential genomic biomarker to predict response to ICI treatment in patients with mRCC, mainly in second- and later treatment lines, but the existence of conflicting data in the literature highlights an important bias in the studies and the need for additional clinical validation in large, prospective trials.
Collapse
Affiliation(s)
- André Paternò Castello Dias Carneiro
- Latin American Cooperative Oncology Group (LACOG), Porto Alegre, Brazil
- Hospital Israelita Albert Einstein, São Paulo, Brazil
- Centro Paulista de Oncologia/Oncoclínicas, São Paulo, Brazil
| | - Fernando Sabino Marques Monteiro
- Latin American Cooperative Oncology Group (LACOG), Porto Alegre, Brazil
- Hospital Santa Lucia, Brasilia, Brazil
- Hospital Universitário de Brasília, Brasilia, Brazil
| | - Andrey Soares
- Latin American Cooperative Oncology Group (LACOG), Porto Alegre, Brazil
- Hospital Israelita Albert Einstein, São Paulo, Brazil
- Centro Paulista de Oncologia/Oncoclínicas, São Paulo, Brazil
| |
Collapse
|
99
|
Chabanon RM, Morel D, Eychenne T, Colmet-Daage L, Bajrami I, Dorvault N, Garrido M, Meisenberg C, Lamb A, Ngo C, Hopkins SR, Roumeliotis TI, Jouny S, Hénon C, Kawai-Kawachi A, Astier C, Konde A, Del Nery E, Massard C, Pettitt SJ, Margueron R, Choudhary JS, Almouzni G, Soria JC, Deutsch E, Downs JA, Lord CJ, Postel-Vinay S. PBRM1 Deficiency Confers Synthetic Lethality to DNA Repair Inhibitors in Cancer. Cancer Res 2021; 81:2888-2902. [PMID: 33888468 DOI: 10.1158/0008-5472.can-21-0628] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/17/2021] [Accepted: 03/19/2021] [Indexed: 11/16/2022]
Abstract
Inactivation of Polybromo 1 (PBRM1), a specific subunit of the PBAF chromatin remodeling complex, occurs frequently in cancer, including 40% of clear cell renal cell carcinomas (ccRCC). To identify novel therapeutic approaches to targeting PBRM1-defective cancers, we used a series of orthogonal functional genomic screens that identified PARP and ATR inhibitors as being synthetic lethal with PBRM1 deficiency. The PBRM1/PARP inhibitor synthetic lethality was recapitulated using several clinical PARP inhibitors in a series of in vitro model systems and in vivo in a xenograft model of ccRCC. In the absence of exogenous DNA damage, PBRM1-defective cells exhibited elevated levels of replication stress, micronuclei, and R-loops. PARP inhibitor exposure exacerbated these phenotypes. Quantitative mass spectrometry revealed that multiple R-loop processing factors were downregulated in PBRM1-defective tumor cells. Exogenous expression of the R-loop resolution enzyme RNase H1 reversed the sensitivity of PBRM1-deficient cells to PARP inhibitors, suggesting that excessive levels of R-loops could be a cause of this synthetic lethality. PARP and ATR inhibitors also induced cyclic GMP-AMP synthase/stimulator of interferon genes (cGAS/STING) innate immune signaling in PBRM1-defective tumor cells. Overall, these findings provide the preclinical basis for using PARP inhibitors in PBRM1-defective cancers. SIGNIFICANCE: This study demonstrates that PARP and ATR inhibitors are synthetic lethal with the loss of PBRM1, a PBAF-specific subunit, thus providing the rationale for assessing these inhibitors in patients with PBRM1-defective cancer. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/81/11/2888/F1.large.jpg.
Collapse
MESH Headings
- Animals
- Apoptosis
- Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/metabolism
- Carcinoma, Non-Small-Cell Lung/pathology
- Carcinoma, Renal Cell/drug therapy
- Carcinoma, Renal Cell/genetics
- Carcinoma, Renal Cell/metabolism
- Carcinoma, Renal Cell/pathology
- Cell Proliferation
- DNA Repair
- DNA-Binding Proteins/deficiency
- Female
- Gene Expression Regulation, Neoplastic/drug effects
- Humans
- Kidney Neoplasms/drug therapy
- Kidney Neoplasms/genetics
- Kidney Neoplasms/metabolism
- Kidney Neoplasms/pathology
- Lung Neoplasms/drug therapy
- Lung Neoplasms/genetics
- Lung Neoplasms/metabolism
- Lung Neoplasms/pathology
- Mice
- Mice, Inbred NOD
- Mice, SCID
- Poly(ADP-ribose) Polymerase Inhibitors/pharmacology
- Synthetic Lethal Mutations
- Transcription Factors/deficiency
- Tumor Cells, Cultured
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Roman M Chabanon
- ATIP-Avenir group, Inserm Unit U981, Gustave Roussy, Villejuif, France
- The CRUK Gene Function Laboratory and Breast Cancer Now Toby Robins Breast Cancer Research Centre, The Institute of Cancer Research, London, United Kingdom
| | - Daphné Morel
- ATIP-Avenir group, Inserm Unit U981, Gustave Roussy, Villejuif, France
- Université Paris Saclay, Université Paris-Sud, Faculté de Médicine, Le Kremlin Bicêtre, France
| | - Thomas Eychenne
- ATIP-Avenir group, Inserm Unit U981, Gustave Roussy, Villejuif, France
| | - Léo Colmet-Daage
- ATIP-Avenir group, Inserm Unit U981, Gustave Roussy, Villejuif, France
| | - Ilirjana Bajrami
- The CRUK Gene Function Laboratory and Breast Cancer Now Toby Robins Breast Cancer Research Centre, The Institute of Cancer Research, London, United Kingdom
| | - Nicolas Dorvault
- ATIP-Avenir group, Inserm Unit U981, Gustave Roussy, Villejuif, France
| | - Marlène Garrido
- ATIP-Avenir group, Inserm Unit U981, Gustave Roussy, Villejuif, France
| | - Cornelia Meisenberg
- Epigenetics and Genome Stability Team, The Institute of Cancer Research, London, United Kingdom
| | | | - Carine Ngo
- ATIP-Avenir group, Inserm Unit U981, Gustave Roussy, Villejuif, France
| | - Suzanna R Hopkins
- Epigenetics and Genome Stability Team, The Institute of Cancer Research, London, United Kingdom
| | | | - Samuel Jouny
- The CRUK Gene Function Laboratory and Breast Cancer Now Toby Robins Breast Cancer Research Centre, The Institute of Cancer Research, London, United Kingdom
| | - Clémence Hénon
- ATIP-Avenir group, Inserm Unit U981, Gustave Roussy, Villejuif, France
| | | | - Clémence Astier
- ATIP-Avenir group, Inserm Unit U981, Gustave Roussy, Villejuif, France
| | - Asha Konde
- The CRUK Gene Function Laboratory and Breast Cancer Now Toby Robins Breast Cancer Research Centre, The Institute of Cancer Research, London, United Kingdom
| | - Elaine Del Nery
- Institut Curie, PSL Research University, Department of Translational Research, The Biophenics High-Content Screening Laboratory, Cell and Tissue Imaging Facility (PICT-IBiSA), Paris, France
| | | | - Stephen J Pettitt
- The CRUK Gene Function Laboratory and Breast Cancer Now Toby Robins Breast Cancer Research Centre, The Institute of Cancer Research, London, United Kingdom
| | - Raphaël Margueron
- Institut Curie, PSL Research University, INSERM Unit U934, CNRS UMR 3215, Paris, France
| | - Jyoti S Choudhary
- Functional Proteomics Team, The Institute of Cancer Research, London, United Kingdom
| | - Geneviève Almouzni
- Institut Curie, PSL Research University, CNRS, UMR 3664, Equipe Labellisée Ligue contre le Cancer, Paris, France
- Sorbonne Universités, UPMC Université Paris-VI, CNRS, UMR3664, Paris, France
| | - Jean-Charles Soria
- Université Paris Saclay, Université Paris-Sud, Faculté de Médicine, Le Kremlin Bicêtre, France
| | - Eric Deutsch
- Université Paris Saclay, Université Paris-Sud, Faculté de Médicine, Le Kremlin Bicêtre, France
- INSERM UMR1030 Molecular Radiotherapy and Therapeutic Innovations, Gustave Roussy, Villejuif, France
| | - Jessica A Downs
- Epigenetics and Genome Stability Team, The Institute of Cancer Research, London, United Kingdom
| | - Christopher J Lord
- The CRUK Gene Function Laboratory and Breast Cancer Now Toby Robins Breast Cancer Research Centre, The Institute of Cancer Research, London, United Kingdom.
| | - Sophie Postel-Vinay
- ATIP-Avenir group, Inserm Unit U981, Gustave Roussy, Villejuif, France.
- Université Paris Saclay, Université Paris-Sud, Faculté de Médicine, Le Kremlin Bicêtre, France
- Drug Development Department, DITEP, Gustave Roussy, Villejuif, France
| |
Collapse
|
100
|
Lee CH, DiNatale RG, Chowell D, Krishna C, Makarov V, Valero C, Vuong L, Lee M, Weiss K, Hoen D, Morris L, Reznik E, Murray S, Kotecha R, Voss MH, Carlo MI, Feldman D, Sachdev P, Adachi Y, Minoshima Y, Matsui J, Funahashi Y, Nomoto K, Hakimi AA, Motzer RJ, Chan TA. High Response Rate and Durability Driven by HLA Genetic Diversity in Patients with Kidney Cancer Treated with Lenvatinib and Pembrolizumab. Mol Cancer Res 2021; 19:1510-1521. [PMID: 34039647 DOI: 10.1158/1541-7786.mcr-21-0053] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 03/20/2021] [Accepted: 05/20/2021] [Indexed: 11/16/2022]
Abstract
Immune checkpoint blockade (ICB) therapy has substantially improved the outcomes of patients with many types of cancers, including renal cell carcinoma (RCC). Initially studied as monotherapy, immunotherapy-based combination regimens have improved the clinical benefit achieved by ICB monotherapy and have revolutionized RCC treatment. While biomarkers like PD-L1 and tumor mutational burden (TMB) are FDA approved as biomarkers for ICB monotherapy, there are no known biomarkers for combination immunotherapies. Here, we describe the clinical outcomes and genomic determinants of response from a phase Ib/II clinical trial on patients with advanced RCC evaluating the efficacy of lenvatinib, a multi-kinase inhibitor mainly targeting VEGFR and FGFR plus pembrolizumab, an anti-PD1 immunotherapy. Concurrent treatment with lenvatinib and pembrolizumab resulted in an objective response rate of 79% (19/24) and tumor shrinkage in 96% (23/24) of patients. While tumor mutational burden (TMB) did not predict for clinical benefit, germline HLA-I diversity strongly impacted treatment efficacy. Specifically, HLA-I evolutionary divergence (HED), which measures the breadth of a patient's immunopeptidome, was associated with both improved clinical benefit and durability of response. Our results identify lenvatinib plus pembrolizumab as a highly active treatment strategy in RCC and reveal HLA-I diversity as a critical determinant of efficacy for this combination. HED also predicted better survival in a separate cohort of patients with RCC following therapy with anti-PD-1-based combination therapy. IMPLICATIONS: These findings have substantial implications for RCC therapy and for understanding immunogenetic mechanisms of efficacy and warrants further investigation.
Collapse
Affiliation(s)
- Chung-Han Lee
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.
| | - Renzo G DiNatale
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Urology, Memorial Sloan Kettering Cancer Center, New York, New York.,Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Diego Chowell
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, New York.,Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Chirag Krishna
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York.,Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Vladimir Makarov
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York.,Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Cristina Valero
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, New York.,Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Lynda Vuong
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, New York.,Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Mark Lee
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, New York.,Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Kate Weiss
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, New York.,Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Doug Hoen
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, New York.,Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Luc Morris
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, New York.,Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ed Reznik
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, New York.,Computational Oncology, Department of Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York.,Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Samuel Murray
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ritesh Kotecha
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Martin H Voss
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Maria I Carlo
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Darren Feldman
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | | | | | | | | | | | | | - A Ari Hakimi
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Urology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Robert J Motzer
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Timothy A Chan
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, New York. .,Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York.,Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, Ohio
| |
Collapse
|