1
|
Zhang X, Zhao J, Yin X, Liang J, Wang Y, Zheng L, Tan P, Lin Y, Xu N, Zhu S, Chen J, Zhao J, Hu X, Pan X, Nie L, Zhang M, Chen Y, Zhang Y, Liu H, Dai J, Wang Z, Liu H, Ni Y, Rupp NJ, Moch H, Sheng X, Gong K, Liu X, Chen Z, He Z, Wang Y, Xu L, Liu M, Zhou H, Tang B, Huang R, Wei Q, Li X, Liu J, Yao J, Liao B, Liu Z, Shen P, Chen N, Zeng H, Sun G. Comprehensive molecular profiling of FH-deficient renal cell carcinoma identifies molecular subtypes and potential therapeutic targets. Nat Commun 2025; 16:4398. [PMID: 40355427 PMCID: PMC12069531 DOI: 10.1038/s41467-025-59513-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 04/25/2025] [Indexed: 05/14/2025] Open
Abstract
Fumarate hydratase-deficient renal cell carcinoma (FH-deficient RCC) is a rare yet highly lethal kidney cancer. To deepen our understanding of FH-deficient RCC, we conduct a comprehensive integrated genomic study. We analyze the association of FH alteration patterns with tumor heterogeneity and develop a CpG site-specific methylation signature for precise identification of FH-deficient RCC. Transcriptomic analysis unveils three distinctive molecular subtypes characterized by enrichment of immune/Angiogenic/Stromal (C1), WNT/Notch/MAPK (C2), and proliferation/stemness (C3) pathways, respectively. Tumors in C1 derive the most substantial survival benefit from a combination of immune checkpoint blockade (ICB) and anti-angiogenic therapy. Tumors in C2 display moderate response to this therapeutic approach. In contrast, tumors in C3 exhibit an unfavorable response to anti-angiogenic monotherapy and its combination with ICB. These findings contribute to a profound understanding of the aggressive nature of FH-deficient RCC, offering insights into potential precision medicine approaches for disease management.
Collapse
Affiliation(s)
- Xingming Zhang
- Department of Urology, Institute of Urology, Sichuan Clinical Research Center for kidney and urologic diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Junjie Zhao
- Department of Urology, Institute of Urology, Sichuan Clinical Research Center for kidney and urologic diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaoxue Yin
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Jiayu Liang
- Department of Urology, Institute of Urology, Sichuan Clinical Research Center for kidney and urologic diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Yongquan Wang
- Department of Urology, Southwest Hospital, Army Medical University, Chongqing, China
| | - Linmao Zheng
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Ping Tan
- Department of Urology, Institute of Urology, Sichuan Clinical Research Center for kidney and urologic diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Yifei Lin
- Department of Urology, Institute of Urology, Sichuan Clinical Research Center for kidney and urologic diseases, West China Hospital, Sichuan University, Chengdu, China
- Department of Urology, Medical Device Regulatory Research and Evaluation Center, West China Hospital, Sichuan University, Chengdu, China
| | - Nanwei Xu
- Department of Urology, Institute of Urology, Sichuan Clinical Research Center for kidney and urologic diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Sha Zhu
- Department of Urology, Institute of Urology, Sichuan Clinical Research Center for kidney and urologic diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Junru Chen
- Department of Urology, Institute of Urology, Sichuan Clinical Research Center for kidney and urologic diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Jinge Zhao
- Department of Urology, Institute of Urology, Sichuan Clinical Research Center for kidney and urologic diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Xu Hu
- Department of Urology, Institute of Urology, Sichuan Clinical Research Center for kidney and urologic diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Xiuyi Pan
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Ling Nie
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Mengni Zhang
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Yuntian Chen
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Yaowen Zhang
- Department of Urology, Institute of Urology, Sichuan Clinical Research Center for kidney and urologic diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Haoyang Liu
- Department of Urology, Institute of Urology, Sichuan Clinical Research Center for kidney and urologic diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Jindong Dai
- Department of Urology, Institute of Urology, Sichuan Clinical Research Center for kidney and urologic diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Zhipeng Wang
- Department of Urology, Institute of Urology, Sichuan Clinical Research Center for kidney and urologic diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Haolin Liu
- Department of Urology, Institute of Urology, Sichuan Clinical Research Center for kidney and urologic diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Yuchao Ni
- Department of Urology, Institute of Urology, Sichuan Clinical Research Center for kidney and urologic diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Niels J Rupp
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
- Faculty of Medicine, Zurich, CH-8006, University of Zurich, Zurich, Switzerland
| | - Holger Moch
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
- Faculty of Medicine, Zurich, CH-8006, University of Zurich, Zurich, Switzerland
| | - Xinan Sheng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Genitourinary Oncology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Kan Gong
- Department of Urology, Peking University First Hospital, Beijing, China
| | - Xiaodong Liu
- Department of Urology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Zhibin Chen
- Department of Urology, The First People's Hospital of Neijiang, Neijiang, China
| | - Zhengyu He
- Department of Urology, Yaan People's Hospital, Yaan, China
| | - Yaodong Wang
- Department of Urology, Mianyang Central Hospital, Mianyang, China
| | - Lijing Xu
- Department of Urology, Institute of Urology, Sichuan Clinical Research Center for kidney and urologic diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Mingsheng Liu
- The Second Ward of Urology, Affiliated Qujing Hospital of Kunming Medical University, Qujing, China
| | - Hongqing Zhou
- The Second Ward of Urology, Affiliated Qujing Hospital of Kunming Medical University, Qujing, China
| | - Bo Tang
- Department of Urology, Institute of Urology, Sichuan Clinical Research Center for kidney and urologic diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Rui Huang
- Department of Nuclear medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Qiang Wei
- Department of Urology, Institute of Urology, Sichuan Clinical Research Center for kidney and urologic diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Xiang Li
- Department of Urology, Institute of Urology, Sichuan Clinical Research Center for kidney and urologic diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Jiyan Liu
- Department of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Jin Yao
- Department of Urology, Southwest Hospital, Army Medical University, Chongqing, China
| | - Banghua Liao
- Department of Urology, Institute of Urology, Sichuan Clinical Research Center for kidney and urologic diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Zhenhua Liu
- Department of Urology, Institute of Urology, Sichuan Clinical Research Center for kidney and urologic diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Pengfei Shen
- Department of Urology, Institute of Urology, Sichuan Clinical Research Center for kidney and urologic diseases, West China Hospital, Sichuan University, Chengdu, China.
| | - Ni Chen
- Department of Urology, Institute of Urology, Sichuan Clinical Research Center for kidney and urologic diseases, West China Hospital, Sichuan University, Chengdu, China.
| | - Hao Zeng
- Department of Urology, Institute of Urology, Sichuan Clinical Research Center for kidney and urologic diseases, West China Hospital, Sichuan University, Chengdu, China.
| | - Guangxi Sun
- Department of Urology, Institute of Urology, Sichuan Clinical Research Center for kidney and urologic diseases, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
2
|
Vento J, Zhang T, Kapur P, Hammers H, Brugarolas J, Qin Q. Systemic Treatment of Locally Advanced or Metastatic Non-Clear Cell Renal Cell Carcinoma. Cancers (Basel) 2025; 17:1527. [PMID: 40361453 PMCID: PMC12071997 DOI: 10.3390/cancers17091527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2025] [Revised: 04/24/2025] [Accepted: 04/28/2025] [Indexed: 05/15/2025] Open
Abstract
Non-clear cell renal cell carcinoma (nccRCC) represents a heterogenous group of malignancies with varying degrees of clinical aggressiveness and response to different systemic therapies. As the characterization of subtypes of nccRCC continues to evolve, it is important to understand the evidence around systemic treatments used in advanced or metastatic stages of specific subtypes. Here, we review the literature on systemic therapies in nccRCC, with a focus on prospective trials that included patients with papillary renal cell carcinoma (RCC), chromophobe RCC, RCC not further classified/unclassified RCC, translocation RCC, collecting duct RCC, and renal medullary carcinoma. We also review emerging treatments for other molecularly defined subtypes of this disease.
Collapse
Affiliation(s)
- Joseph Vento
- Division of Hematology and Oncology, Department of Internal Medicine, University of Texas Southwestern, Dallas, TX 75235, USA
| | - Tian Zhang
- Division of Hematology and Oncology, Department of Internal Medicine, University of Texas Southwestern, Dallas, TX 75235, USA
| | - Payal Kapur
- Department of Pathology, University of Texas Southwestern, Dallas, TX 75235, USA
| | - Hans Hammers
- Division of Hematology and Oncology, Department of Internal Medicine, University of Texas Southwestern, Dallas, TX 75235, USA
| | - James Brugarolas
- Division of Hematology and Oncology, Department of Internal Medicine, University of Texas Southwestern, Dallas, TX 75235, USA
| | - Qian Qin
- Division of Hematology and Oncology, Department of Internal Medicine, University of Texas Southwestern, Dallas, TX 75235, USA
| |
Collapse
|
3
|
Giri VK, Zaemes J. The selection of targeted therapies for relapsed or refractory advanced renal cell carcinoma. Expert Rev Anticancer Ther 2025; 25:337-349. [PMID: 39998618 DOI: 10.1080/14737140.2025.2468765] [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: 01/07/2025] [Revised: 02/03/2025] [Accepted: 02/14/2025] [Indexed: 02/27/2025]
Abstract
INTRODUCTION Advancements in immunotherapy and angiogenesis-targeted therapies have transformed the upfront treatment of renal cell carcinoma (RCC). However, long-term prognoses for patients with unresectable and metastatic disease often remain limited, with the majority experiencing progression after exposure to front-line therapy. In most cases of relapsed or refractory (R/R) disease after prior exposure to an immune checkpoint inhibitor (ICI), there is no role for ICI-rechallenge. Therefore, treatment of R/R RCC relies on the appropriate selection of therapies targeting growth pathways dependent on vascular endothelial growth factor (VEGF) or hypoxia-inducible factor (HIF). AREAS COVERED This review article summarizes the current landscape of targeted therapies for use in second-line or later-line settings for the treatment of clear cell and non-clear cell RCC. Novel therapeutic strategies currently in development are also discussed. EXPERT OPINION The treatment of R/R RCC primarily consists of inhibition of VEGF, HIF, and mTOR pathways, and the selection of a specific agent depends on the histologic subtype of the tumor, the prior lines of therapy chosen, and patient co-morbidities. Future tumor-based and circulating biomarker research might one day enable the identification of transcriptional signatures that could predict a response to immune, angiogenesis, or HIF-based therapies.
Collapse
Affiliation(s)
- Vinay K Giri
- Department of Medicine, Division of Medical Oncology, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Jacob Zaemes
- Department of Medicine, Division of Medical Oncology, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, MA, USA
| |
Collapse
|
4
|
Marletta S, Marcolini L, Caliò A, Pedron S, Antonini P, Martelli FM, Stefanizzi L, Martignoni G. Stimulator of interferon genes (STING) immunohistochemical expression in fumarate hydratase-deficient renal cell carcinoma: biological and potential predictive implications. Virchows Arch 2025:10.1007/s00428-025-04041-5. [PMID: 39899047 DOI: 10.1007/s00428-025-04041-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 01/13/2025] [Accepted: 01/23/2025] [Indexed: 02/04/2025]
Abstract
Fumarate hydratase (FH)-deficient renal cell carcinoma is an aggressive neoplasm driven by inactivating mutations of the FH gene, which cause metabolites like S-(2-succinyl)cysteine (2SC) to accumulate and trigger cascades supporting malignant transformation. Although in preclinical models the c-GAS-STING pathway is activated by fumarate metabolites, its role in humans has not been explored yet. Eleven FH-deficient renal cell carcinomas, including primary neoplasms and metastases, were retrieved and evaluated for clinical-pathological features and immunohistochemical expression of FH, 2SC (commercially available), and STING. The in-house collection accounted for 0.2% of the 2011-2023 renal cell carcinomas cohort (5/2210). Eight-on-ten cases with available follow-up behaved aggressively (local recurrence/distant metastases). All tumors revealed FH staining loss and strong and diffuse 2SC immunolabeling. At least focal STING expression was detected in most primary tumors (9/11, 82%), often (78%) in a wide percentage of cells (≥ 30%). Notably, significant STING expression was observed in all but two aggressive renal neoplasms, with one of the remaining showing increased staining in its hepatic localization, and in 86% (6/7) of neoplasms significantly expressing PD-L1. In our series, (i) FH-deficient renal cell carcinoma represents 0.2% of in-house cases; (ii) combining FH loss and positive 2SC staining now commercially available is useful in primary and secondary tumors, supporting this latter marker's safe routine adoption; and (iii) a significant STING labeling (≥ 30%) in most of the samples, especially in those behaving aggressively and expressing PD-L1, provides novel insights regarding the molecular basis of FH-deficient renal cell carcinomas, proposing STING as a potential predictive marker.
Collapse
Affiliation(s)
- S Marletta
- Department of Diagnostic and Public Health, Section of Pathology, University of Verona, Largo L. Scuro 10, 37134, Verona, Italy
- Division of Pathology, Humanitas Istituto Clinico Catanese, Catania, Italy
| | - L Marcolini
- Department of Pathology, Pederzoli Hospital, Peschiera del Garda, Italy
| | - A Caliò
- Department of Diagnostic and Public Health, Section of Pathology, University of Verona, Largo L. Scuro 10, 37134, Verona, Italy
| | - S Pedron
- Department of Diagnostic and Public Health, Section of Pathology, University of Verona, Largo L. Scuro 10, 37134, Verona, Italy
| | - P Antonini
- Department of Diagnostic and Public Health, Section of Pathology, University of Verona, Largo L. Scuro 10, 37134, Verona, Italy
| | - F M Martelli
- Department of Diagnostic and Public Health, Section of Pathology, University of Verona, Largo L. Scuro 10, 37134, Verona, Italy
- Department of Pathology, Pederzoli Hospital, Peschiera del Garda, Italy
| | - L Stefanizzi
- Department of Pathology, Pederzoli Hospital, Peschiera del Garda, Italy
| | - G Martignoni
- Department of Diagnostic and Public Health, Section of Pathology, University of Verona, Largo L. Scuro 10, 37134, Verona, Italy.
- Department of Pathology, Pederzoli Hospital, Peschiera del Garda, Italy.
| |
Collapse
|
5
|
Zhang Y, Chen J, Wang X, Wang H, Chen X, Hong J, Fang H. Effectiveness of systemic treatments for advanced non-clear cell renal cell carcinoma: a systematic review and meta-analysis. Front Oncol 2024; 14:1478245. [PMID: 39743997 PMCID: PMC11688242 DOI: 10.3389/fonc.2024.1478245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 12/03/2024] [Indexed: 01/04/2025] Open
Abstract
Background Non-clear cell renal cell carcinoma (nccRCC) represents a heterogeneous group of malignancies with substantial differences in morphology, genetic profiles, clinical behavior, and prognosis. Optimal treatment for nccRCC remains unclear, largely extrapolated from evidence available for clear cell renal cell carcinoma (ccRCC). This study aimed to compare the efficacy of current mainstream drug treatments for nccRCC to provide clinical treatment guidance for advanced cases. Methods We systematically searched PubMed, Embase, and Cochrane databases for trials published up to January 2, 2024, including controlled and single-arm trials. Primary outcomes included overall response rate (ORR), disease control rate (DCR), progression-free survival (PFS), and overall survival (OS). Results We selected six randomized controlled trials (RCTs) comparing mammalian target of rapamycin inhibitors (mTORi) with vascular endothelial growth factor receptor tyrosine kinase inhibitors (VEGFR-TKIs). These trials included four first-line and two second-line studies, with a total of 398 advanced nccRCC patients. Pooled results showed that VEGFR-TKIs significantly improved PFS compared to mTORi in first-line treatment (relative risk [RR] = 1.387; 95% confidence interval [CI]: 1.04-1.85; p = 0.026). In a single-arm meta-analysis, we included 22 VEGFR-TKI trials, three mTORi trials, 12 immune checkpoint inhibitor (ICI) therapies, five chemotherapy trials, and 10 combination therapy trials. The pooled ORR ranged from 6% (95% CI: 0-16%) to 36% (95% CI: 27-44%), and the pooled DCR ranged from 54% (95% CI: 50-58%) to 81% (95% CI: 70-91%). Subgroup analysis of ICI showed a higher ORR in the PD-L1 positive group compared to the PD-L1 negative group (RR = 3.044; 95% CI: 1.623-5.709; p = 0.001). Conclusion This systematic review and meta-analysis demonstrate that VEGFR-TKIs improve PFS in first-line treatment compared to mTORi. The single-arm meta-analysis suggest that combination therapies with different mechanisms result in better ORR and DCR. Furthermore, PD-L1 positive patients showed significantly better therapeutic responses with ICI treatment than PD-L1 negative patients.
Collapse
Affiliation(s)
- Yaping Zhang
- Department of Oncology, Affliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou, China
| | - Jian Chen
- Department of GCP, Affliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou, China
| | - Xiaoyan Wang
- Department of Oncology, Affliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou, China
| | - Hui Wang
- Department of Oncology, Affliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou, China
| | - Xiaoli Chen
- Department of Oncology, Affliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou, China
| | - Jianfeng Hong
- Department of Oncology, Affliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou, China
| | - Hongming Fang
- Department of Oncology, Affliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou, China
| |
Collapse
|
6
|
Pezzicoli G, Musci V, Ciciriello F, Salonne F, Cafforio P, Lionetti N, Ragno A, Rizzo M. Genomic profiling and molecular characterization of non-clear cell renal cell carcinoma: a narrative review from a clinical perspective. Ther Adv Med Oncol 2024; 16:17588359241298500. [PMID: 39563719 PMCID: PMC11574901 DOI: 10.1177/17588359241298500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 10/23/2024] [Indexed: 11/21/2024] Open
Abstract
While the clear-cell renal cell carcinoma (ccRCC) treatment has undergone several paradigm shifts in recent years, the non-clear cell renal cell carcinoma (nccRCC) therapeutic approach has yet to be extensively investigated and improved. The WHO 2022 classification of renal neoplasms redefined the most common nccRCC subtypes (papillary and chromophobe RCC) and introduced the molecularly defined RCC class, which is a first step in the direction of better molecular profiling of nccRCC. We reviewed the literature data on known genomic alterations of clinical interest in nccRCC and discussed their potential role in guiding therapeutic choices in each nccRCC entity. Among the alterations discussed, we focused on the ones that could be treated with already available drugs, such as MET-driven papillary RCC, mechanistic target of rapamycin altered chromophobe RCC, anaplastic lymphoma kinase-rearranged RCC, and fumarate-hydratase deficient RCC. Furthermore, we focused on the currently ongoing clinical trials and further evidence for all the other entities, such as SMARCB1-deficient RCC, TFE3 and transcription factorEB (TFEB)-altered RCC, and Elongin C (ELOC)-mutated RCC. The vast heterogeneity of nccRCC does not allow a one-size-fits-all solution; therefore, molecular characterization is the path toward effective therapies and fully personalized medicine for these entities.
Collapse
Affiliation(s)
- Gaetano Pezzicoli
- Department of Interdisciplinary Medicine, University of Bari "Aldo Moro," Bari, Italy
| | - Vittoria Musci
- Department of Interdisciplinary Medicine, University of Bari "Aldo Moro," Bari, Italy
| | - Federica Ciciriello
- Department of Interdisciplinary Medicine, University of Bari "Aldo Moro," Bari, Italy
| | - Francesco Salonne
- Department of Interdisciplinary Medicine, University of Bari "Aldo Moro," Bari, Italy
| | - Paola Cafforio
- Department of Interdisciplinary Medicine, University of Bari "Aldo Moro," Bari, Italy
| | - Nicoletta Lionetti
- Department of Interdisciplinary Medicine, University of Bari "Aldo Moro," Bari, Italy
| | - Anna Ragno
- Medical Oncology Unit, Azienda Ospedaliera Universitaria Consorziale, Policlinico di Bari, Bari, Italy
| | - Mimma Rizzo
- Department of Interdisciplinary Medicine, University of Bari "Aldo Moro," Bari, Italy
- Medical Oncology Unit, Azienda Ospedaliera Universitaria Consorziale, Policlinico di Bari, piazza G. Cesare 11, Bari 70124, Italy
| |
Collapse
|
7
|
Noronha KJ, Lucas KN, Paradkar S, Edmonds J, Friedman S, Murray MA, Liu S, Sajed DP, Sachs C, Spurrier J, Raponi M, Liang J, Zeng H, Sundaram RK, Shuch B, Vasquez JC, Bindra RS. NAPRT Silencing in FH-Deficient Renal Cell Carcinoma Confers Therapeutic Vulnerabilities via NAD+ Depletion. Mol Cancer Res 2024; 22:973-988. [PMID: 38949523 PMCID: PMC11445649 DOI: 10.1158/1541-7786.mcr-23-1003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 05/10/2024] [Accepted: 06/21/2024] [Indexed: 07/02/2024]
Abstract
Hereditary leiomyomatosis and renal cell carcinoma (HLRCC) is caused by loss of function mutations in fumarate hydratase (FH) and results in an aggressive subtype of renal cell carcinoma with limited treatment options. Loss of FH leads to accumulation of fumarate, an oncometabolite that disrupts multiple cellular processes and drives tumor progression. High levels of fumarate inhibit alpha ketoglutarate-dependent dioxygenases, including the ten-eleven translocation (TET) enzymes, and can lead to global DNA hypermethylation. Here, we report patterns of hypermethylation in FH-mutant cell lines and tumor samples are associated with the silencing of nicotinate phosphoribosyl transferase (NAPRT), a rate-limiting enzyme in the Preiss-Handler pathway of NAD+ biosynthesis, in a subset of HLRCC cases. NAPRT is hypermethylated at a CpG island in the promoter in cell line models and patient samples, resulting in loss of NAPRT expression. We find that FH-deficient RCC models with loss of NAPRT expression, as well as other oncometabolite-producing cancer models that silence NAPRT, are extremely sensitive to nicotinamide phosphoribosyl transferase inhibitors (NAMPTi). NAPRT silencing was also associated with synergistic tumor cell killing with PARP inhibitors and NAMPTis, which was associated with effects on PAR-mediated DNA repair. Overall, our findings indicate that NAPRT silencing can be targeted in oncometabolite-producing cancers and elucidates how oncometabolite-associated hypermethylation can impact diverse cellular processes and lead to therapeutically relevant vulnerabilities in cancer cells. Implications: NAPRT is a novel biomarker for targeting NAD+ metabolism in FH-deficient HLRCCs with NAMPTis alone and targeting DNA repair processes with the combination of NAMPTis and PARP inhibitors.
Collapse
Affiliation(s)
- Katelyn J. Noronha
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut.
| | - Karlie N. Lucas
- Department of Therapeutic Radiology, Yale University, New Haven, Connecticut.
| | - Sateja Paradkar
- Department of Experimental Pathology, Yale University, New Haven, Connecticut.
| | - Joseph Edmonds
- Department of Therapeutic Radiology, Yale University, New Haven, Connecticut.
| | - Sam Friedman
- Department of Therapeutic Radiology, Yale University, New Haven, Connecticut.
| | - Matthew A. Murray
- Department of Experimental Pathology, Yale University, New Haven, Connecticut.
| | - Samantha Liu
- Department of Therapeutic Radiology, Yale University, New Haven, Connecticut.
| | - Dipti P. Sajed
- Department of Pathology, University of California Los Angeles, Los Angeles, California.
| | - Chana Sachs
- Department of Pathology, University of California Los Angeles, Los Angeles, California.
| | | | | | - Jiayu Liang
- Department of Urology, West China Hospital, Sichuan University, Chengdu, P.R. China.
| | - Hao Zeng
- Department of Urology, West China Hospital, Sichuan University, Chengdu, P.R. China.
| | - Ranjini K. Sundaram
- Department of Therapeutic Radiology, Yale University, New Haven, Connecticut.
| | - Brian Shuch
- Institute of Urologic Oncology, Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, California.
| | - Juan C. Vasquez
- Department of Pediatric Hematology and Oncology, Yale University, New Haven, Connecticut.
| | - Ranjit S. Bindra
- Department of Therapeutic Radiology, Yale University, New Haven, Connecticut.
| |
Collapse
|
8
|
Caliò A, Marletta S, Stefanizzi L, Marcolini L, Rotellini M, Serio G, Bariani E, Vicentini C, Pedron S, Martelli FM, Antonini P, Brunelli M, Martignoni G. Comparison of Primary and Metastatic Fumarate Hydratase-Deficient Renal Cell Carcinomas Documents Morphologic Divergence and Potential Diagnostic Pitfall With Peritoneal Mesothelioma. Mod Pathol 2024; 37:100561. [PMID: 38996839 DOI: 10.1016/j.modpat.2024.100561] [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: 02/23/2024] [Revised: 06/05/2024] [Accepted: 07/01/2024] [Indexed: 07/14/2024]
Abstract
Fumarate hydratase (FH)-deficient renal cell carcinomas are rare neoplasms characterized by wide morphologic heterogeneity and pathogenetic mutations in the FH gene. They often show aggressive behavior with rapid diffusion to distant organs, so novel therapeutic scenarios have been explored, including EGFR inhibitors and PD-L1 expression for targeted immunotherapy. Herein, we investigated a series of 11 primary FH-deficient renal cell carcinomas and 7 distant metastases to evaluate tumor heterogeneity even in metastatic sites and estimate the specific spread rates to various organs. Furthermore, the tumors were tested for immunohistochemical PD-L1 expression and EGFR mutations. Most metastatic cases involved the abdominal lymph nodes (4/7; 57%), followed by the peritoneum (3/7; 42%), the liver (2/7; 29%), and the lungs (1/7; 14%). Six metastatic localizations were histologically documented, revealing a morphologic heterogeneous architecture often differing from that of the corresponding primary renal tumor. Peritoneal involvement morphologically resembled a benign reactive mesothelial process or primary peritoneal mesothelioma, thus advocating to perform an accurate immunohistochemical panel, including PAX8 and FH, to reach a proper diagnosis. A pure low-grade succinate dehydrogenase-looking primary FH-deficient renal cell carcinoma was also recorded. As for therapy, significant PD-L1 labeling was found in 60% of primary renal tumors, whereas none of them carried pathogenetic EGFR mutations. Our data show that FH-deficient renal cell carcinoma may be morphologically heterogeneous in metastases as well, which involve the lymph nodes, the liver, and the peritoneum more frequently than other renal tumors. Due to the high frequency of this latter (42%), pathologists should always be concerned about ruling out mesothelial-derived mimickers, and the occurrence of rarer, primary, low-grade-looking types. Finally, contrary to EGFR mutations, PD-L1 expression could be a possible predictive biomarker for the therapy of these tumors.
Collapse
Affiliation(s)
- Anna Caliò
- Section of Pathology, Department of Diagnostic and Public Health, University of Verona, Verona, Italy
| | - Stefano Marletta
- Section of Pathology, Department of Diagnostic and Public Health, University of Verona, Verona, Italy; Division of Pathology, Humanitas Istituto Clinico Catanese, Catania, Italy
| | - Lavinia Stefanizzi
- Department of Pathology and Laboratory Medicine, Pederzoli Hospital, Peschiera, Verona, Italy
| | - Lisa Marcolini
- Department of Pathology and Laboratory Medicine, Pederzoli Hospital, Peschiera, Verona, Italy
| | - Matteo Rotellini
- Anatomia Patologica Massa Carrara Azienda Toscana Nord Ovest, Italy
| | - Gabriella Serio
- Pathology Unit, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari, Italy
| | - Elena Bariani
- Unit of Anatomic Pathology, Department of Oncology, Bellaria Hospital, Bologna, Italy
| | - Caterina Vicentini
- Department of Pathology and Laboratory Medicine, Pederzoli Hospital, Peschiera, Verona, Italy
| | - Serena Pedron
- Section of Pathology, Department of Diagnostic and Public Health, University of Verona, Verona, Italy
| | - Filippo M Martelli
- Department of Pathology and Laboratory Medicine, Pederzoli Hospital, Peschiera, Verona, Italy
| | - Pietro Antonini
- Section of Pathology, Department of Diagnostic and Public Health, University of Verona, Verona, Italy
| | - Matteo Brunelli
- Section of Pathology, Department of Diagnostic and Public Health, University of Verona, Verona, Italy
| | - Guido Martignoni
- Section of Pathology, Department of Diagnostic and Public Health, University of Verona, Verona, Italy; Department of Pathology and Laboratory Medicine, Pederzoli Hospital, Peschiera, Verona, Italy.
| |
Collapse
|
9
|
Powles T, Albiges L, Bex A, Comperat E, Grünwald V, Kanesvaran R, Kitamura H, McKay R, Porta C, Procopio G, Schmidinger M, Suarez C, Teoh J, de Velasco G, Young M, Gillessen S. Renal cell carcinoma: ESMO Clinical Practice Guideline for diagnosis, treatment and follow-up. Ann Oncol 2024; 35:692-706. [PMID: 38788900 DOI: 10.1016/j.annonc.2024.05.537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 05/03/2024] [Accepted: 05/13/2024] [Indexed: 05/26/2024] Open
Affiliation(s)
- T Powles
- Barts Cancer Institute, Department of Medical Oncology, Queen Mary University of London and Royal Free London NHS Foundation Trust, London, UK
| | - L Albiges
- Université Paris Saclay, Institut Gustave Roussy, Villejuif, France
| | - A Bex
- Specialist Centre for Kidney Cancer, Royal Free London NHS Foundation Trust, London; Division of Surgery and Interventional Science, University College London, London, UK; Department of Urology, The Netherlands Cancer Institute, Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - E Comperat
- Department of Pathology, Medical University of Vienna, General Hospital of Vienna, Vienna, Austria
| | - V Grünwald
- Interdisciplinary Genitourinary Oncology, West German Cancer Center Clinic for Internal Medicine and Clinic for Urology, University Hospital Essen, Essen, Germany
| | - R Kanesvaran
- Division of Medical Oncology, National Cancer Centre, Singapore, Singapore
| | - H Kitamura
- Department of Urology, Faculty of Medicine, University of Toyama, Toyama, Japan
| | - R McKay
- Department of Medicine and Urology, University of California San Diego, La Jolla, USA
| | - C Porta
- Interdisciplinary Department of Medicine, University of Bari 'A. Moro', Bari; Division of Medical Oncology, A.O.U. Consorziale Policlinico di Bari, Bari
| | - G Procopio
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - M Schmidinger
- Department of Urology, Medical University of Vienna, Vienna, Austria
| | - C Suarez
- Medical Oncology, Vall d'Hebron Institute of Oncology (VHIO), Hospital Universitari Vall d'Hebron, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - J Teoh
- S. H. Ho Urology Centre, Department of Surgery, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - G de Velasco
- Instituto de Investigación i+12 and Departamento de Oncología Médica, Hospital University 12 de Octubre, Madrid, Spain
| | - M Young
- Barts Cancer Institute, Department of Medical Oncology, Queen Mary University of London and Royal Free London NHS Foundation Trust, London, UK; Barts Cancer Institute, Department of Experimental Cancer Medicine, Queen Mary University of London, London, UK
| | - S Gillessen
- Oncology Institute of Southern Switzerland (IOSI), Ente Ospedaliero Cantonale (EOC), Bellinzona; Università della Svizzera Italiana, Lugano, Switzerland
| |
Collapse
|
10
|
Liu B, Zhou H, Tan L, Siu KTH, Guan XY. Exploring treatment options in cancer: Tumor treatment strategies. Signal Transduct Target Ther 2024; 9:175. [PMID: 39013849 PMCID: PMC11252281 DOI: 10.1038/s41392-024-01856-7] [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: 01/23/2024] [Revised: 04/24/2024] [Accepted: 04/29/2024] [Indexed: 07/18/2024] Open
Abstract
Traditional therapeutic approaches such as chemotherapy and radiation therapy have burdened cancer patients with onerous physical and psychological challenges. Encouragingly, the landscape of tumor treatment has undergone a comprehensive and remarkable transformation. Emerging as fervently pursued modalities are small molecule targeted agents, antibody-drug conjugates (ADCs), cell-based therapies, and gene therapy. These cutting-edge treatment modalities not only afford personalized and precise tumor targeting, but also provide patients with enhanced therapeutic comfort and the potential to impede disease progression. Nonetheless, it is acknowledged that these therapeutic strategies still harbour untapped potential for further advancement. Gaining a comprehensive understanding of the merits and limitations of these treatment modalities holds the promise of offering novel perspectives for clinical practice and foundational research endeavours. In this review, we discussed the different treatment modalities, including small molecule targeted drugs, peptide drugs, antibody drugs, cell therapy, and gene therapy. It will provide a detailed explanation of each method, addressing their status of development, clinical challenges, and potential solutions. The aim is to assist clinicians and researchers in gaining a deeper understanding of these diverse treatment options, enabling them to carry out effective treatment and advance their research more efficiently.
Collapse
Affiliation(s)
- Beilei Liu
- Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong, China
- State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong, China
| | - Hongyu Zhou
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong, China
| | - Licheng Tan
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong, China
| | - Kin To Hugo Siu
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong, China
| | - Xin-Yuan Guan
- Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China.
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong, China.
- State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong, China.
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou, China.
- MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangzhou, China.
| |
Collapse
|
11
|
Kim M, Shim HS, Kim S, Lee IH, Kim J, Yoon S, Kim HD, Park I, Jeong JH, Yoo C, Cheon J, Kim IH, Lee J, Hong SH, Park S, Jung HA, Kim JW, Kim HJ, Cha Y, Lim SM, Kim HS, Lee CK, Kim JH, Chun SH, Yun J, Park SY, Lee HS, Cho YM, Nam SJ, Na K, Yoon SO, Lee A, Jang KT, Yun H, Lee S, Kim JH, Kim WS. Clinical practice recommendations for the use of next-generation sequencing in patients with solid cancer: a joint report from KSMO and KSP. J Pathol Transl Med 2024; 58:147-164. [PMID: 39026440 PMCID: PMC11261170 DOI: 10.4132/jptm.2023.11.01] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 10/31/2023] [Accepted: 11/01/2023] [Indexed: 07/20/2024] Open
Abstract
In recent years, next-generation sequencing (NGS)-based genetic testing has become crucial in cancer care. While its primary objective is to identify actionable genetic alterations to guide treatment decisions, its scope has broadened to encompass aiding in pathological diagnosis and exploring resistance mechanisms. With the ongoing expansion in NGS application and reliance, a compelling necessity arises for expert consensus on its application in solid cancers. To address this demand, the forthcoming recommendations not only provide pragmatic guidance for the clinical use of NGS but also systematically classify actionable genes based on specific cancer types. Additionally, these recommendations will incorporate expert perspectives on crucial biomarkers, ensuring informed decisions regarding circulating tumor DNA panel testing.
Collapse
Affiliation(s)
- Miso Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Hyo Sup Shim
- Department of Pathology, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Sheehyun Kim
- Department of Genomic Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - In Hee Lee
- Department of Oncology/Hematology, Kyungpook National University Chilgok Hospital, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Jihun Kim
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Shinkyo Yoon
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Hyung-Don Kim
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Inkeun Park
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jae Ho Jeong
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Changhoon Yoo
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jaekyung Cheon
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - In-Ho Kim
- Division of Medical Oncology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jieun Lee
- Division of Medical Oncology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Sook Hee Hong
- Division of Medical Oncology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Sehhoon Park
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hyun Ae Jung
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jin Won Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Han Jo Kim
- Division of Oncology and Hematology, Department of Internal Medicine, Soonchunhyang University Cheonan Hospital, Cheonan, Korea
| | - Yongjun Cha
- Division of Medical Oncology, Center for Colorectal Cancer, National Cancer Center, Goyang, Korea
| | - Sun Min Lim
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Korea
| | - Han Sang Kim
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Korea
| | - Choong-Kun Lee
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Korea
| | - Jee Hung Kim
- Division of Medical Oncology, Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Sang Hoon Chun
- Division of Medical Oncology, Department of Internal Medicine, Bucheon St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jina Yun
- Division of Hematology/Oncology, Department of Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Korea
| | - So Yeon Park
- Department of Pathology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Hye Seung Lee
- Department of Pathology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Yong Mee Cho
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Soo Jeong Nam
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Kiyong Na
- Department of Pathology, Kyung Hee University Hospital, Kyung Hee University College of Medicine, Seoul, Korea
| | - Sun Och Yoon
- Department of Pathology, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Ahwon Lee
- Department of Hospital Pathology, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Kee-Taek Jang
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hongseok Yun
- Department of Genomic Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Sungyoung Lee
- Department of Genomic Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Jee Hyun Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Wan-Seop Kim
- Department of Pathology, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul, Korea
| |
Collapse
|
12
|
Kim M, Shim HS, Kim S, Lee IH, Kim J, Yoon S, Kim HD, Park I, Jeong JH, Yoo C, Cheon J, Kim IH, Lee J, Hong SH, Park S, Jung HA, Kim JW, Kim HJ, Cha Y, Lim SM, Kim HS, Lee CK, Kim JH, Chun SH, Yun J, Park SY, Lee HS, Cho YM, Nam SJ, Na K, Yoon SO, Lee A, Jang KT, Yun H, Lee S, Kim JH, Kim WS. Clinical Practice Recommendations for the Use of Next-Generation Sequencing in Patients with Solid Cancer: A Joint Report from KSMO and KSP. Cancer Res Treat 2024; 56:721-742. [PMID: 38037319 PMCID: PMC11261187 DOI: 10.4143/crt.2023.1043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 11/17/2023] [Indexed: 12/02/2023] Open
Abstract
In recent years, next-generation sequencing (NGS)-based genetic testing has become crucial in cancer care. While its primary objective is to identify actionable genetic alterations to guide treatment decisions, its scope has broadened to encompass aiding in pathological diagnosis and exploring resistance mechanisms. With the ongoing expansion in NGS application and reliance, a compelling necessity arises for expert consensus on its application in solid cancers. To address this demand, the forthcoming recommendations not only provide pragmatic guidance for the clinical use of NGS but also systematically classify actionable genes based on specific cancer types. Additionally, these recommendations will incorporate expert perspectives on crucial biomarkers, ensuring informed decisions regarding circulating tumor DNA panel testing.
Collapse
Affiliation(s)
- Miso Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Hyo Sup Shim
- Department of Pathology, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Sheehyun Kim
- Department of Genomic Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - In Hee Lee
- Department of Oncology/Hematology, Kyungpook National University Chilgok Hospital, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Jihun Kim
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Shinkyo Yoon
- Department of Oncology,Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Hyung-Don Kim
- Department of Oncology,Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Inkeun Park
- Department of Oncology,Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jae Ho Jeong
- Department of Oncology,Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Changhoon Yoo
- Department of Oncology,Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jaekyung Cheon
- Department of Oncology,Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - In-Ho Kim
- Division of Medical Oncology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jieun Lee
- Division of Medical Oncology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Sook Hee Hong
- Division of Medical Oncology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Sehhoon Park
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hyun Ae Jung
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jin Won Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Han Jo Kim
- Division of Oncology and Hematology, Department of Internal Medicine, Soonchunhyang University Cheonan Hospital, Cheonan, Korea
| | - Yongjun Cha
- Division of Medical Oncology, Center for Colorectal Cancer, National Cancer Center, Goyang, Korea
| | - Sun Min Lim
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Korea
| | - Han Sang Kim
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Korea
| | - Choong-kun Lee
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Korea
| | - Jee Hung Kim
- Division of Medical Oncology, Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Sang Hoon Chun
- Division of Medical Oncology, Department of Internal Medicine, Bucheon St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jina Yun
- Division of Hematology/Oncology, Department of Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Korea
| | - So Yeon Park
- Department of Pathology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Hye Seung Lee
- Department of Pathology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Yong Mee Cho
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Soo Jeong Nam
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Kiyong Na
- Department of Pathology, Kyung Hee University Hospital, Kyung Hee University College of Medicine, Seoul, Korea
| | - Sun Och Yoon
- Department of Pathology, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Ahwon Lee
- Department of Hospital Pathology, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Kee-Taek Jang
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hongseok Yun
- Department of Genomic Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Sungyoung Lee
- Department of Genomic Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Jee Hyun Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Wan-Seop Kim
- Department of Pathology, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul, Korea
| |
Collapse
|
13
|
Chen J, Hu X, Zhao J, Yin X, Zheng L, Guo J, Chen J, Wang Y, Sheng X, Dong H, Liu X, Zhang X, Liang J, Liu H, Yao J, Liu J, Shen Y, Chen Z, He Z, Wang Y, Chen N, Nie L, Zhang M, Pan X, Chen Y, Liu H, Zhang Y, Tang Y, Zhu S, Zhao J, Dai J, Wang Z, Zeng Y, Wang Z, Huang H, Liu Z, Shen P, Zeng H, Sun G. Memory/Active T-Cell Activation Is Associated with Immunotherapeutic Response in Fumarate Hydratase-Deficient Renal Cell Carcinoma. Clin Cancer Res 2024; 30:2571-2581. [PMID: 38512114 PMCID: PMC11145163 DOI: 10.1158/1078-0432.ccr-23-2760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 11/22/2023] [Accepted: 03/19/2024] [Indexed: 03/22/2024]
Abstract
PURPOSE Fumarate hydratase-deficient renal cell carcinoma (FH-deficient RCC) is a rare and lethal subtype of kidney cancer. However, the optimal treatments and molecular correlates of benefits for FH-deficient RCC are currently lacking. EXPERIMENTAL DESIGN A total of 91 patients with FH-deficient RCC from 15 medical centers between 2009 and 2022 were enrolled in this study. Genomic and bulk RNA-sequencing (RNA-seq) were performed on 88 and 45 untreated FH-deficient RCCs, respectively. Single-cell RNA-seq was performed to identify biomarkers for treatment response. Main outcomes included disease-free survival (DFS) for localized patients, objective response rate (ORR), progression-free survival (PFS), and overall survival (OS) for patients with metastasis. RESULTS In the localized setting, we found that a cell-cycle progression signature enabled to predict disease progression. In the metastatic setting, first-line immune checkpoint inhibitor plus tyrosine kinase inhibitor (ICI+TKI) combination therapy showed satisfactory safety and was associated with a higher ORR (43.2% vs. 5.6%), apparently superior PFS (median PFS, 17.3 vs. 9.6 months, P = 0.016) and OS (median OS, not reached vs. 25.7 months, P = 0.005) over TKI monotherapy. Bulk and single-cell RNA-seq data revealed an enrichment of memory and effect T cells in responders to ICI plus TKI combination therapy. Furthermore, we identified a signature of memory and effect T cells that was associated with the effectiveness of ICI plus TKI combination therapy. CONCLUSIONS ICI plus TKI combination therapy may represent a promising treatment option for metastatic FH-deficient RCC. A memory/active T-cell-derived signature is associated with the efficacy of ICI+TKI but necessitates further validation.
Collapse
Affiliation(s)
- Junru Chen
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Xu Hu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Junjie Zhao
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaoxue Yin
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Linmao Zheng
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Jingjing Guo
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, China
| | - Jianhui Chen
- Department of Urology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Yongquan Wang
- Department of Urology, Southwest Hospital, Army Medical University, Chongqing, China
| | - Xinan Sheng
- Department of Genitourinary Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Haiying Dong
- Department of Urology, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, China
| | - Xiaodong Liu
- Department of Urology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xingming Zhang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Jiayu Liang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Haolin Liu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Jin Yao
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Jiyan Liu
- Department of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yali Shen
- Department of Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Zhibin Chen
- Department of Urology, The First People's Hospital of Neijiang, Neijiang, China
| | - Zhengyu He
- Department of Urology, Yaan People's Hospital, Yaan, China
| | - Yaodong Wang
- Department of Urology, Mianyang Central Hospital, Mianyang, China
| | - Ni Chen
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Ling Nie
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Mengni Zhang
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Xiuyi Pan
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Yuntian Chen
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Haoyang Liu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Yaowen Zhang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Yanfeng Tang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Sha Zhu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Jinge Zhao
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Jindong Dai
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Zilin Wang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Yuhao Zeng
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Zhipeng Wang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Haojie Huang
- Institute of Urological Cancer Research, Zhejiang University School of Medicine, The First Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - Zhenhua Liu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Pengfei Shen
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Hao Zeng
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Guangxi Sun
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
14
|
Soares A, Monteiro FSM, da Trindade KM, Silva AGE, Cardoso APG, Sasse AD, Fay AP, Carneiro APCD, Alencar Junior AM, de Andrade Mota AC, Santucci B, da Motta Girardi D, Herchenhorn D, Araújo DV, Jardim DL, Bastos DA, Rosa DR, Schutz FA, Kater FR, da Silva Marinho F, Maluf FC, de Oliveira FNG, Vidigal F, Morbeck IAP, Rinck Júnior JA, Costa LAGA, Maia MCDF, Zereu M, Freitas MRP, Dias MSF, Tariki MS, Muniz P, Beato PMM, Lages PSM, Velho PI, de Carvalho RS, Mariano RC, de Araújo Cavallero SR, Oliveira TM, Souza VC, Smaletz O, de Cássio Zequi S. Advanced renal cell carcinoma management: the Latin American Cooperative Oncology Group (LACOG) and the Latin American Renal Cancer Group (LARCG) consensus update. J Cancer Res Clin Oncol 2024; 150:183. [PMID: 38594593 PMCID: PMC11003910 DOI: 10.1007/s00432-024-05663-z] [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: 12/27/2023] [Accepted: 02/22/2024] [Indexed: 04/11/2024]
Abstract
PURPOSE Renal cell carcinoma is an aggressive disease with a high mortality rate. Management has drastically changed with the new era of immunotherapy, and novel strategies are being developed; however, identifying systemic treatments is still challenging. This paper presents an update of the expert panel consensus from the Latin American Cooperative Oncology Group and the Latin American Renal Cancer Group on advanced renal cell carcinoma management in Brazil. METHODS A panel of 34 oncologists and experts in renal cell carcinoma discussed and voted on the best options for managing advanced disease in Brazil, including systemic treatment of early and metastatic renal cell carcinoma as well as nonclear cell tumours. The results were compared with the literature and graded according to the level of evidence. RESULTS Adjuvant treatments benefit patients with a high risk of recurrence after surgery, and the agents used are pembrolizumab and sunitinib, with a preference for pembrolizumab. Neoadjuvant treatment is exceptional, even in initially unresectable cases. First-line treatment is mainly based on tyrosine kinase inhibitors (TKIs) and immune checkpoint inhibitors (ICIs); the choice of treatment is based on the International Metastatic Database Consortium (IMCD) risk score. Patients at favourable risk receive ICIs in combination with TKIs. Patients classified as intermediate or poor risk receive ICIs, without preference for ICI + ICIs or ICI + TKIs. Data on nonclear cell renal cancer treatment are limited. Active surveillance has a place in treating favourable-risk patients. Either denosumab or zoledronic acid can be used for treating metastatic bone disease. CONCLUSION Immunotherapy and targeted therapy are the standards of care for advanced disease. The utilization and sequencing of these therapeutic agents hinge upon individual risk scores and responses to previous treatments. This consensus reflects a commitment to informed decision-making, drawn from professional expertise and evidence in the medical literature.
Collapse
Affiliation(s)
- Andrey Soares
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil.
- Hospital Israelita Albert Einstein, São Paulo, SP, Brazil.
- Centro Paulista de Oncologia/Oncoclínicas, São Paulo, SP, Brazil.
| | - Fernando Sabino Marques Monteiro
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- Hospital Sírio-Libanês, Brasília, DF, Brazil
| | - Karine Martins da Trindade
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- Oncologia D'Or, Fortaleza, CE, Brazil
| | - Adriano Gonçalves E Silva
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- Instituto do Câncer e Transplante de Curitiba/PR (ICTr Curitiba), Curitiba, PR, Brazil
| | - Ana Paula Garcia Cardoso
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- Hospital Israelita Albert Einstein, São Paulo, SP, Brazil
| | - André Deeke Sasse
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- Grupo SONHE de Campinas, Campinas, SP, Brazil
| | - André P Fay
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- Escola de Medicina da Pontifícia, Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil
| | - André Paternò Castello Dias Carneiro
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- Hospital Israelita Albert Einstein, São Paulo, SP, Brazil
- Hospital Municipal Vila Santa Catarina, São Paulo, SP, Brazil
| | - Antonio Machado Alencar Junior
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- Hospital São Domingos, São Luís, MA, Brazil
- Dasa Oncologia, Brasília, DF, Brazil
- Hospital Universitário da Universidade Federal do Maranhão (UFMA), São Luís, MA, Brazil
| | - Augusto César de Andrade Mota
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- Clínica AMO-DASA, Feira de Santana, BA, Brazil
| | - Bruno Santucci
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- Instituto Paulista de Cancerologia, São Paulo, SP, Brazil
| | - Daniel da Motta Girardi
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- Hospital Sírio-Libanês, Brasília, DF, Brazil
| | - Daniel Herchenhorn
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- Oncologia D'Or, Rio de Janeiro, RJ, Brazil
| | - Daniel Vilarim Araújo
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- Hospital de Base de São José do Rio Preto/SP, São José do Rio Preto, São Paulo, SP, Brazil
| | - Denis Leonardo Jardim
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- Grupo Oncoclínicas, São Paulo, São Paulo, SP, Brazil
| | - Diogo Assed Bastos
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- Hospital Sirio-Libanês de São Paulo, São Paulo, SP, Brazil
| | - Diogo Rodrigues Rosa
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- Grupo Oncoclínicas, Rio de Janeiro, RJ, Brazil
| | - Fabio A Schutz
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- Beneficência Portuguesa de São Paulo, São Paulo, SP, Brazil
| | - Fábio Roberto Kater
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- Beneficência Portuguesa de São Paulo, São Paulo, SP, Brazil
| | - Felipe da Silva Marinho
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- Grupo Oncoclínicas, Recife, PE, Brazil
| | - Fernando Cotait Maluf
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- Hospital Israelita Albert Einstein, São Paulo, SP, Brazil
- Beneficência Portuguesa de São Paulo, São Paulo, SP, Brazil
| | - Fernando Nunes Galvão de Oliveira
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- Grupo Oncoclínicas, Salvador, BA, Brazil
| | - Fernando Vidigal
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- Dasa Oncologia, Brasília, DF, Brazil
| | - Igor Alexandre Protzner Morbeck
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- Grupo Oncoclínicas, Brasília, DF, Brazil
| | - Jose Augusto Rinck Júnior
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- AC Camargo Cancer Center, São Paulo, SP, Brazil
| | - Leonardo Atem G A Costa
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- Oncologia D'Or, Fortaleza, CE, Brazil
| | - Manuel Caitano Dias Ferreira Maia
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- Hospital do Câncer Porto Dias, Belém, PA, Brazil
| | - Manuela Zereu
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- Santa Casa de Misericórdia de Porto Alegre, Porto Alegre, RS, Brazil
| | - Marcelo Roberto Pereira Freitas
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- Centro Especializado de Oncologia de Florianópolis, Florianópolis, SC, Brazil
| | - Mariane Sousa Fontes Dias
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- Grupo Oncoclínicas, Rio de Janeiro, RJ, Brazil
| | - Milena Shizue Tariki
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- AC Camargo Cancer Center, São Paulo, SP, Brazil
| | - Pamela Muniz
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- Grupo Oncoclínicas, São Paulo, São Paulo, SP, Brazil
- Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, Brazil
| | - Patrícia Medeiros Milhomem Beato
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- Hospital Amaral Carvalho, Jaú, SP, Brazil
| | - Paulo Sérgio Moraes Lages
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- Grupo Oncoclínicas, Brasília, DF, Brazil
| | - Pedro Isaacsson Velho
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- Hospital Moinhos de Vento, Porto Alegre, RS, Brazil
- Johns Hopkins University, Baltimore, MD, USA
| | - Ricardo Saraiva de Carvalho
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- Beneficência Portuguesa de São Paulo, São Paulo, SP, Brazil
| | - Rodrigo Coutinho Mariano
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- Beneficência Portuguesa de São Paulo, São Paulo, SP, Brazil
| | - Sandro Roberto de Araújo Cavallero
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- Hospital Adventista de Belém, Belém, PA, Brazil
| | - Thiago Martins Oliveira
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- Hospital São Rafael, Salvador, BA, Brazil
| | - Vinicius Carrera Souza
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- Instituto D'Or de Ensino e Pesquisa, Salvador, BA, Brazil
| | - Oren Smaletz
- Latin American Cooperative Oncology Group, Genitourinary Group (LACOG-GU), Av. Brigadeiro Faria Lima, Vila Olímpia, São Paulo, SP, 4300, Brazil
- Hospital Israelita Albert Einstein, São Paulo, SP, Brazil
| | - Stênio de Cássio Zequi
- AC Camargo Cancer Center, São Paulo, SP, Brazil
- National Institute for Science and Technology in Oncogenomics and Therapeutic Innovation, AC Camargo Cancer Center, São Paulo, SP, Brazil
| |
Collapse
|
15
|
Wilson NR, Acikgoz Y, Hasanov E. Advances in non-clear cell renal cell carcinoma management: From heterogeneous biology to treatment options. Int J Cancer 2024; 154:947-961. [PMID: 37823185 DOI: 10.1002/ijc.34756] [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: 06/01/2023] [Revised: 09/15/2023] [Accepted: 09/19/2023] [Indexed: 10/13/2023]
Abstract
Non-clear cell renal cell carcinoma (nccRCC) makes up nearly one quarter of all RCC subtypes, commonly impacts younger patients, and is often metastatic at presentation. Compared to clear-cell RCC (ccRCC), nccRCC typically has a worse prognosis in the metastatic setting, with overall survival durations that are ~10 months shorter. The nccRCC consists of a wide range of different histological subtypes, the majority of which are composed of papillary, chromophobe, renal medullary carcinoma, translocation RCC, collecting duct carcinoma and unclassified RCC. Most clinical trials have either excluded or only included small numbers of patients with nccRCC; owing to the lack of prospective studies focusing on this population, data on response rates and survival outcomes are lacking. NccRCC treatment is a nascent field with various therapeutic modalities and combinations under investigation, often based on data extrapolated from therapeutic studies in ccRCC. We herein review the use and outcomes of cytotoxic chemotherapy, various combination modalities of tyrosine kinase inhibitors and immune checkpoint inhibitors, and targeted agents. We discuss active ongoing clinical trials for patients with nccRCC and future directions in the treatment of this rare disease. Historically, treatment for nccRCC has been adopted from the standard of care for patients with ccRCC, although these treatments are less effective in the nccRCC population. As we begin to understand the underlying biology of these tumors, clinical trials have been able to slowly accrue and include more patients with various subtypes of nccRCC. There remains much room for improvement in this area of need, but there is hope on the horizon.
Collapse
Affiliation(s)
- Nathaniel R Wilson
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan; Michigan Medicine, Ann Arbor, Michigan, USA
| | - Yusuf Acikgoz
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA
| | - Elshad Hasanov
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA
- Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA
| |
Collapse
|
16
|
Chen YB. Update on Selected High-grade Renal Cell Carcinomas of the Kidney: FH-deficient, ALK-rearranged, and Medullary Carcinomas. Adv Anat Pathol 2024; 31:118-125. [PMID: 38145398 PMCID: PMC11232671 DOI: 10.1097/pap.0000000000000426] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2023]
Abstract
High-grade renal cell carcinoma (RCC), often diagnosed at advanced stages, significantly contributes to renal cancer-related mortality. This review explores the progress in understanding specific subtypes of high-grade RCC, namely fumarate hydratase (FH)-deficient RCC, anaplastic lymphoma kinase (ALK)-rearranged RCC, and SMARCB1-deficient renal medullary carcinoma, all of which are now recognized as molecularly defined entities in the WHO classification system (2022). While these entities each exhibit a morphologic spectrum that overlaps with other high-grade RCC, ancillary tools developed based on their distinctive molecular alterations can help establish a specific diagnosis, underscoring the importance of integrating molecular findings into diagnostic paradigms. It is important to exclude these specific tumor types in cases with similar morphologic spectrum before rendering a diagnosis of high-grade papillary RCC, collecting duct carcinoma, or RCC, NOS. Several gray areas exist within the spectrum of high-grade uncommon types of RCC, necessitating continued research to enhance diagnostic precision and therapeutic options.
Collapse
Affiliation(s)
- Ying-Bei Chen
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
17
|
Jang A, Hobeika CS, Gupta S. Papillary Renal Cell Carcinoma: Current Evidence and Future Directions. KIDNEY CANCER 2024; 8:61-79. [PMID: 40027140 PMCID: PMC11870658 DOI: 10.3233/kca-230027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Papillary renal cell carcinoma (pRCC) comprises 15-20% of all patients with renal cell carcinoma (RCC). Although in the localized setting where pRCC appears to have better outcomes than clear cell RCC (ccRCC), patients with metastatic pRCC have significantly worse outcomes than patients with metastatic ccRCC. Because of the overall rarity of pRCC, there have been less research and clinical trials devoted to this subtype. Therefore, treatment of pRCC has generally been extrapolated from approved therapies for ccRCC. Recent data shows promise with newer tyrosine kinase inhibitors, and there is emerging evidence on their combination with immune checkpoint inhibitors. However, more dedicated clinical trials to pRCC are urgently needed, as response rates and outcomes still lag behind ccRCC. This review summarizes the pathophysiology, genetic features, the evolution of treatment approaches since the systemic cytokine era, and current challenges of managing pRCC.
Collapse
Affiliation(s)
- Albert Jang
- Section of Solid Tumor Oncology, University Hospitals Seidman Cancer Center, Case Comprehensive Cancer Center, Cleveland, OH, USA
| | - Charbel S. Hobeika
- Department of Internal Medicine, Cleveland Clinic Fairview Hospital, Cleveland, OH, USA
| | - Shilpa Gupta
- Cleveland Clinic Taussig Cancer Institute, Case Comprehensive Cancer Center, Cleveland, OH, USA
| |
Collapse
|
18
|
Naik P, Dudipala H, Chen YW, Rose B, Bagrodia A, McKay RR. The incidence, pathogenesis, and management of non-clear cell renal cell carcinoma. Ther Adv Urol 2024; 16:17562872241232578. [PMID: 38434237 PMCID: PMC10906063 DOI: 10.1177/17562872241232578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Accepted: 01/18/2024] [Indexed: 03/05/2024] Open
Abstract
Renal cell carcinoma (RCC) is the most common type of kidney cancer and is divided into two distinct subtypes, clear cell renal cell carcinoma (ccRCC) and non-clear cell renal cell carcinoma (nccRCC). Although many treatments exist for RCC, these are largely based on clinical trials performed in ccRCC and there are limited studies on the management of nccRCC. Non-clear cell RCC consists of multiple histological subtypes: papillary, chromophobe, translocation, medullary, collecting duct, unclassified, and other rare histologies. Due to variations in pathogenesis and therapeutic response, therapy should be tailored to specific variant histologies. For patients with localized nccRCC, surgical resection remains the gold standard. In the metastatic setting, the standard of care has yet to be clearly defined, and most guidelines recommend clinical trial participation. General therapeutic options include immunotherapy, either as monotherapy or in combination, targeted therapies such as vascular endothelial growth factor tyrosine kinase inhibitors and MET inhibitors, and chemotherapy in certain subtypes. Here we present a review of the incidence and pathogenesis of the various subtypes, as well as available clinical data to support therapeutic recommendations for these subtypes. We also highlight currently available clinical trials in nccRCC and future directions in investigating novel treatment modalities tailored to patients with variant histology.
Collapse
Affiliation(s)
- Priyanka Naik
- Undergraduate Studies, University of California, San Diego, La Jolla, CA, USA
| | - Harshitha Dudipala
- Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, 72 E Concord St, Boston, MA 02118, USA
| | - Yu-Wei Chen
- Department of Hematology and Oncology, Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - Brent Rose
- Department of Radiation Oncology, Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - Aditya Bagrodia
- Department of Urology, University of California, San Diego, La Jolla, CA, USA
| | - Rana R. McKay
- Department of Hematology and Oncology, Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|
19
|
Samar MR, Khan W, Rashid YA, Mohammad ATV. Unmasking the enigma: A case of Fumarate Hydratase-deficient renal cell carcinoma. Int J Surg Case Rep 2023; 113:109054. [PMID: 37992668 DOI: 10.1016/j.ijscr.2023.109054] [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: 10/16/2023] [Revised: 11/09/2023] [Accepted: 11/10/2023] [Indexed: 11/24/2023] Open
Abstract
INTRODUCTION Fumarate Hydratase-deficient-renal cell carcinoma (FH-dRCC) is an uncommon and extremely aggressive variant of renal cell carcinoma (RCC) resulting from inactivating mutations in the tumor suppressor gene, fumarate hydratase (FH). CASE PRESENTATION We report a case of a young male who presented with the complaint of painless hematuria. Upon workup, he was found to have a renal mass with bony metastases. The histopathology was consistent with renal cell carcinoma with features of FH-deficient variant. Germline testing showed a pathogenic mutation in the FH gene. He was started on a treatment combination of Pembrolizumab and Axitinib along with Zoledronate for bone metastasis. His response to the combination therapy was short with early progression of disease. He was switched to a second-line treatment Bevacizumab and Erlotinib, which achieved significant disease response. DISCUSSION Systemic therapy is the mainstay of treatment for metastatic disease. Although the novel agents approved for other subsets of RCC have been used, the responses are grim. There is no consensus on the sequence of further lines of treatment for FH-dRCC and is based on the physician's choice, availability of the drugs, cost, toxicity, and tolerance of the patient. CONCLUSION This case report emphasizes the occurrence, presentation, management and prognosis of FH-dRCC, which is an aggressive entity, presenting at a young age with early distant metastases, not diagnosed appropriately due to its poorly characterized cytologic features. Being an infrequent neoplasm, it is an area that warrants oncological exploration to improve outcomes in these individuals. The combination of Erlotinib and Bevacizumab provides promising outcomes in terms of progression-free survival.
Collapse
Affiliation(s)
- Mirza Rameez Samar
- Department of Medical Oncology, The Aga Khan University Hospital, Pakistan.
| | - Wajiha Khan
- Department of Medicine And Surgery, Dow University of Health Sciences, Karachi, Pakistan
| | | | | |
Collapse
|
20
|
Valcarcel-Jimenez L, Frezza C. Fumarate hydratase (FH) and cancer: a paradigm of oncometabolism. Br J Cancer 2023; 129:1546-1557. [PMID: 37689804 PMCID: PMC10645937 DOI: 10.1038/s41416-023-02412-w] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 08/10/2023] [Accepted: 08/18/2023] [Indexed: 09/11/2023] Open
Abstract
Fumarate hydratase (FH) is an enzyme of the Tricarboxylic Acid (TCA) cycle whose mutations lead to hereditary and sporadic forms of cancer. Although more than twenty years have passed since its discovery as the leading cause of the cancer syndrome Hereditary leiomyomatosis and Renal Cell Carcinoma (HLRCC), it is still unclear how the loss of FH causes cancer in a tissue-specific manner and with such aggressive behaviour. It has been shown that FH loss, via the accumulation of FH substrate fumarate, activates a series of oncogenic cascades whose contribution to transformation is still under investigation. In this review, we will summarise these recent findings in an integrated fashion and put forward the case that understanding the biology of FH and how its mutations promote transformation will be vital to establish novel paradigms of oncometabolism.
Collapse
Affiliation(s)
- Lorea Valcarcel-Jimenez
- Department of Biochemistry and Molecular Biology, Faculty of Science and Technology, UPV/EHU, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain.
| | - Christian Frezza
- University of Cologne, Faculty of Mathematics and Natural Sciences, Institute of Genetics, Cluster of Excellence Cellular Stress Responses in Aging-associated Diseases (CECAD), Cologne, Germany.
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Cluster of Excellence Cellular Stress Responses in Aging-associated Diseases (CECAD), Cologne, Germany.
| |
Collapse
|
21
|
Msaouel P, Genovese G, Tannir NM. Renal Cell Carcinoma of Variant Histology: Biology and Therapies. Hematol Oncol Clin North Am 2023; 37:977-992. [PMID: 37244822 PMCID: PMC11608423 DOI: 10.1016/j.hoc.2023.04.019] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
The term variant histology renal cell carcinomas (vhRCCs), also known as non-clear cell RCCs, refers to a diverse group of malignancies with distinct biologic and therapeutic considerations. The management of vhRCC subtypes is often based on extrapolating results from the more common clear cell RCC studies or basket trials that are not specific to each histology. The unique management of each vhRCC subtype necessitates accurate pathologic diagnosis and dedicated research efforts. Herein, we discuss tailored recommendations for each vhRCC histology informed by ongoing research and clinical experience.
Collapse
Affiliation(s)
- Pavlos Msaouel
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Translational Molecular Pathology, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA; David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas, MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Giannicola Genovese
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas, MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Genomic Medicine, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA; TRACTION Platform, Division of Therapeutic Discoveries, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Nizar M Tannir
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
22
|
Mimma R, Anna C, Matteo B, Gaetano P, Carlo G, Guido M, Camillo P. Clinico-pathological implications of the 2022 WHO Renal Cell Carcinoma classification. Cancer Treat Rev 2023; 116:102558. [PMID: 37060647 DOI: 10.1016/j.ctrv.2023.102558] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 04/03/2023] [Accepted: 04/07/2023] [Indexed: 04/17/2023]
Abstract
The new WHO classification of urogenital tumours published in 2022, contains significant revisions upon the previous 2016 version regarding Renal Cell Carcinoma (RCC). While the most common histotype remains almost untouched, some of the main novelties concerns papillary RCC and oncocytic neoplasms. The main change is the introduction of a new category of molecularly-defined RCC, which includes TFE3-rearranged RCC, TFEB-rearranged, and TFEB-amplified RCC, FH-deficient RCC, SDH-deficient RCC, ALK-rearranged RCC, ELOC (formerly TCEB1)-mutated RCC, SMARCB1 (INI1)-deficient RCC. In this paper we analyze the current knowledge on emerging entities and molecularly-defined RCC to assess whether the current pathological classification offers the oncologist the possibility of selecting more specific and personalized treatments, from both those currently available, as well as those that will soon be available.
Collapse
Affiliation(s)
- Rizzo Mimma
- Division of Medical Oncology, Azienda Ospedaliero Universitaria Consorziale Policlinico di Bari, Bari, Italy.
| | - Caliò Anna
- Department of Diagnostic and Public Health, Section of Pathology, University of Verona, Italy
| | - Brunelli Matteo
- Department of Diagnostic and Public Health, Section of Pathology, University of Verona, Italy
| | - Pezzicoli Gaetano
- Department of Interdisciplinary Medicine, School of Medicine, University of Bari "A. Moro", Bari, Italy
| | - Ganini Carlo
- Department of Interdisciplinary Medicine, School of Medicine, University of Bari "A. Moro", Bari, Italy
| | - Martignoni Guido
- Department of Diagnostic and Public Health, Section of Pathology, University of Verona, Italy; Department of Pathology, Pederzoli Hospital, Peschiera del Garda, Verona, Italy
| | - Porta Camillo
- 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
|
23
|
Carlo MI. Improving Systemic Therapy for Fumarate Hydratase-deficient Renal Cell Carcinoma. Eur Urol 2023; 83:e113-e114. [PMID: 36682904 DOI: 10.1016/j.eururo.2022.12.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 12/20/2022] [Indexed: 01/21/2023]
Affiliation(s)
- Maria I Carlo
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
24
|
Xu Y, Kong W, Cao M, Wang J, Wang Z, Zheng L, Wu X, Cheng R, He W, Yang B, Dong B, Pan J, Chen Y, Huang J, Jiang C, Zhai W, Li F, Chen R, Zhou X, Wu G, Geng X, Chen J, An H, Yuan Y, Xu T, Chen D, Lin D, Xu L, Huang K, Peng L, Yu Y, Tai S, Qi H, Luo K, Kang X, Wang H, Huang Y, Zhang J, Xue W. Genomic Profiling and Response to Immune Checkpoint Inhibition plus Tyrosine Kinase Inhibition in FH-Deficient Renal Cell Carcinoma. Eur Urol 2023; 83:163-172. [PMID: 35715365 DOI: 10.1016/j.eururo.2022.05.029] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 05/09/2022] [Accepted: 05/28/2022] [Indexed: 02/01/2023]
Abstract
BACKGROUND FH-deficient renal cell carcinoma (RCC) is a rare and exceptionally aggressive RCC subtype. There is currently limited understanding of the molecular alterations, pathogenesis, survival outcomes, and systemic therapy efficacy for this cancer. OBJECTIVE To perform a retrospective multicenter analysis of molecular profiling and clinical outcomes for patients with FH-deficient RCC, with an emphasis on treatment response to first-line immune checkpoint inhibitor plus tyrosine kinase inhibitor (ICI/TKI) versus bevacizumab plus erlotinib (Bev/Erlo) combination therapy in patients with advanced disease. DESIGN, SETTING, AND PARTICIPANTS The study included 77 cases of FH-deficient RCC from 15 centers across China. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS Clinical characteristics, molecular correlates, 18F-fluorodeoxyglucose positron emission tomography/computed tomography imaging, and treatment outcomes were analyzed. RESULTS AND LIMITATIONS A total of 77 patients were identified, including 70 cases with a germline FH alteration (hereditary leiomyomatosis RCC syndrome [HLRCC]-associated RCC) and seven patients with somatic FH loss. Recurrent pathogenic alterations were found in NF2 (six/57, 11%), CDH1 (six/57, 11%), PIK3CA (six/57, 11%), and TP53 (five/57, 8.8%). Sixty-seven patients were evaluable for response to first-line systemic therapy with Bev/Erlo (n = 12), TKI monotherapy (n = 29), or ICI/TKI (n = 26). ICI/TKI combination therapy was associated with more favorable overall survival on systemic treatment (hazard ratio [HR] 0.19, 95% confidence interval [CI] 0.04-0.90) and progression-free survival on first-line therapy (HR 0.22, 95% CI 0.07-0.71) compared to Bev/Erlo combination therapy. The main limitation is the retrospective study design. CONCLUSIONS We described the genomic characteristics of FH-deficient RCC in an Asian population and observed a favorable response to ICI/TKI combinational therapy among patients with advanced disease. PATIENT SUMMARY This real-world study provides evidence supporting the antitumour activity of combining molecular targeted therapy plus immunotherapy for kidney cancer deficient in fumarate hydratase. Further studies are needed to investigate the efficacy of this combination strategy in this rare cancer.
Collapse
Affiliation(s)
- Yunze Xu
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wen Kong
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ming Cao
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jieying Wang
- Clinical Center for Investigation, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zaoyu Wang
- Department of Pathology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Liang Zheng
- Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaoyu Wu
- Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Rongrong Cheng
- Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wei He
- Department of Pathology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Bo Yang
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Baijun Dong
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jiahua Pan
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yonghui Chen
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jiwei Huang
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Chen Jiang
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wei Zhai
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Fangzhou Li
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ruohua Chen
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiang Zhou
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Guangyu Wu
- Department of Radiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaochuan Geng
- Department of Radiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jiasheng Chen
- Department of Urology, Changzhou No. 2 People's Hospital, Changzhou, China
| | - Huimin An
- Department of Urology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yichu Yuan
- Department of Urology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Tianyuan Xu
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Dongning Chen
- Department of Urology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Dengqiang Lin
- Department of Urology, Xiamen Branch, Zhongshan Hospital, Fudan University, Xiamen, China
| | - Lieyu Xu
- Department of Urological Surgery, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, Nanchang, China
| | - Kangbo Huang
- Department of Urology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ling Peng
- Department of Respiratory Disease, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Yanfei Yu
- Department of Urology, Peking University First Hospital, Institute of Urology, Peking University, National Urological Cancer Center, Beijing, China
| | - Shengcheng Tai
- Department of Urology, Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou, China
| | - Honggang Qi
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Kai Luo
- Biobank Department, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaonan Kang
- Biobank Department, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hang Wang
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yiran Huang
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Jin Zhang
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Wei Xue
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
25
|
John A, Spain L, Hamid AA. Navigating the Current Landscape of Non-Clear Cell Renal Cell Carcinoma: A Review of the Literature. Curr Oncol 2023; 30:923-937. [PMID: 36661719 PMCID: PMC9858145 DOI: 10.3390/curroncol30010070] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 12/24/2022] [Accepted: 01/06/2023] [Indexed: 01/11/2023] Open
Abstract
Non-clear cell renal cell carcinoma (nccRCC) is an entity comprised of a heterogeneous constellation of RCC subtypes. Genomic profiling has broadened our understanding of molecular pathogenic mechanisms unique to individual nccRCC subtypes. To date, clinical trials evaluating the use of immunotherapies and targeted therapies have predominantly been conducted in patients with clear cell histology. A comprehensive review of the literature has been undertaken in order to describe molecular pathogenic mechanisms pertaining to each nccRCC subtype, and concisely summarise findings from therapeutic trials conducted in the nccRCC space.
Collapse
Affiliation(s)
- Alexius John
- Department of Medical Oncology, Eastern Health, Melbourne, VIC 3128, Australia
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
| | - Lavinia Spain
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
| | - Anis A. Hamid
- Department of Medical Oncology, Eastern Health, Melbourne, VIC 3128, Australia
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
- Eastern Health Clinical School, Monash University, Melbourne, VIC 3128, Australia
- Department of Surgery, University of Melbourne, Melbourne, VIC 3010, Australia
| |
Collapse
|
26
|
Su WP, Huang YH, Chow NH, Yu YT. Fumarate hydratase-deficient renal cell carcinoma: A case report and literature review. JOURNAL OF CANCER RESEARCH AND PRACTICE 2023. [DOI: 10.4103/ejcrp.ejcrp-d-22-00023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/12/2023] Open
|
27
|
Taylor AS, Skala SL. Tumors masquerading as type 2 papillary renal cell carcinoma: pathologists' ever-expanding differential diagnosis for a heterogeneous group of entities. Urol Oncol 2022; 40:499-511. [PMID: 34116938 DOI: 10.1016/j.urolonc.2021.04.043] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 03/22/2021] [Accepted: 04/28/2021] [Indexed: 02/07/2023]
Abstract
Although papillary renal cell carcinoma has historically been classified as either type 1 or type 2, data from The Cancer Genome Atlas (TCGA) has demonstrated significant genomic heterogeneity in tumors classified as "type 2 papillary renal cell carcinoma" (T2PRCC). Papillary renal cell carcinoma is expected to have a favorable clinical course compared to clear cell renal cell carcinoma (CCRCC). However, tumors with poor outcome more similar to CCRCC were included in the T2PRCC cohort studied by the TCGA. The differential diagnosis for T2PRCC includes a variety of other renal tumors, including aggressive entities such as TFE3 translocation-associated renal cell carcinoma, TFEB-amplified renal cell carcinoma, fumarate hydratase-deficient renal cell carcinoma, high-grade CCRCC, and collecting duct carcinoma. Accurate classification of these tumors is important for prognostication and selection of therapy.
Collapse
Affiliation(s)
- Alexander S Taylor
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI
| | - Stephanie L Skala
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI.
| |
Collapse
|
28
|
Yamana K, Ohashi R, Tomita Y. Contemporary Drug Therapy for Renal Cell Carcinoma- Evidence Accumulation and Histological Implications in Treatment Strategy. Biomedicines 2022; 10:2840. [PMID: 36359359 PMCID: PMC9687261 DOI: 10.3390/biomedicines10112840] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 10/25/2022] [Accepted: 11/02/2022] [Indexed: 09/19/2023] Open
Abstract
Renal cell carcinoma (RCC) is a heterogeneous disease comprising a variety of histological subtypes. Approximately 70-80% of RCC cases are clear cell carcinoma (ccRCC), while the remaining subtypes constitute non-clear cell carcinoma (nccRCC). The medical treatment of RCC has greatly changed in recent years through advances in molecularly targeted therapies and immunotherapies. Most of the novel systemic therapies currently available have been approved based on ccRCC clinical trial data. nccRCC can be subdivided into more than 40 histological subtypes that have distinct clinical, histomorphological, immunohistochemical, and molecular features. These entities are listed as emerging in the 2022 World Health Organization classification. The diagnosis of nccRCC and treatments based on cancer histology and biology remain challenging due to the disease's rarity. We reviewed clinical trials focused on recent discoveries regarding clinicopathological features.
Collapse
Affiliation(s)
- Kazutoshi Yamana
- Department of Urology and Molecular Oncology, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-Dori, Chuo-Ku, Niigata 951-8510, Japan
| | - Riuko Ohashi
- Division of Molecular and Diagnostic Pathology, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-Dori, Chuo-Ku, Niigata 951-8510, Japan
| | - Yoshihiko Tomita
- Department of Urology and Molecular Oncology, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-Dori, Chuo-Ku, Niigata 951-8510, Japan
| |
Collapse
|
29
|
Dong P, Zhang X, Peng Y, Zhang Y, Liu R, Li Y, Pan Q, Wei W, Guo S, Zhang Z, Han H, Zhou F, Liu Y, He L. Genomic Characteristics and Single-Cell Profiles After Immunotherapy in Fumarate Hydratase-Deficient Renal Cell Carcinoma. Clin Cancer Res 2022; 28:4807-4819. [PMID: 36074152 DOI: 10.1158/1078-0432.ccr-22-1279] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 06/23/2022] [Accepted: 09/02/2022] [Indexed: 01/24/2023]
Abstract
PURPOSE Fumarate hydratase-deficient renal cell carcinoma (FHRCC) is highly malignant, but the urgent need for effective treatment remains unmet. We aimed to analyze the genomic characteristics and microenvironment of FHRCC and the cause of heterogeneous response to immune checkpoint inhibitor (ICI)-based treatment at single-cell level. EXPERIMENTAL DESIGN Whole-exome sequencing and IHC staining analyses were performed in 30 advanced FHRCC patients. Single-cell RNA sequencing following ICI-based treatment was conducted in 4 patients. The clinical characteristics, therapeutic effect, and follow-up data were analyzed. RESULTS The median tumor mutation burden was only 0.14 mutations per megabase. IHC staining showed an immune-active tumor microenvironment characterized by extensive CD8+ T-cell infiltration. ATM expression was inversely correlated with percentage of tumor-infiltrating CD8+ T cells. Trajectory analysis indicated gradually upregulated exhausted markers and an increased apoptotic trend of CD8+ T cells despite continuous exposure to ICI-based treatment. ICI-based treatment was associated with improved overall response rate (17.6% vs. 0%, P = 0.046) and disease control rate (DCR; 64.7% vs. 12.5%, P = 0.004) compared with tyrosine kinase inhibitor. Among patients with germline mutation, the ORR (16.7% vs. 0%, P = 0.086) and the DCR (66.7% vs. 14.3%, P = 0.011) were higher after ICI-based treatment. CONCLUSIONS Immune infiltration is frequent in FHRCC. ICI-based treatment is a promising regimen, and treatment response depends on the functional status of tumor-infiltrating lymphocytes. ICI-based treatment cannot reverse the exhaustion of CD8+ T cells in patients with progressive disease, highlighting the need for additional therapeutic strategies.
Collapse
Affiliation(s)
- Pei Dong
- Department of Urology, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Xinyue Zhang
- Department of Radiation Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Yulu Peng
- Department of Urology, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Yijun Zhang
- Department of Pathology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Ruiqi Liu
- Department of Radiation Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Yilin Li
- Department of Radiation Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Qiwen Pan
- Department of Radiation Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Wensu Wei
- Department of Urology, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Shengjie Guo
- Department of Urology, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Zhiling Zhang
- Department of Urology, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Hui Han
- Department of Urology, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Fangjian Zhou
- Department of Urology, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Yang Liu
- Department of Radiation Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Liru He
- Department of Radiation Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| |
Collapse
|
30
|
Sweeney PL, Jang A, Halat SK, Pal SK, Barata PC. Advanced papillary renal cell carcinoma: Epidemiology, genomic drivers, current therapies, and ongoing trials. Cancer Treat Res Commun 2022; 33:100639. [PMID: 36162322 DOI: 10.1016/j.ctarc.2022.100639] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 09/16/2022] [Accepted: 09/21/2022] [Indexed: 12/14/2022]
Abstract
Papillary renal cell carcinoma (PRCC) is the most common subtype of non-clear cell renal cell carcinoma. PRCC can be subdivided into types 1 and 2 based on histology, each associated with different genetic mutations. The MET gene is commonly altered in type 1 PRCC while multiple alterations are involved in type 2 PRCC. PRCC is an aggressive cancer with a predominance in male and black patients and poor prognosis. Due to its rarity, there was a lack of convincing prospective data to guide treatment; hence, therapies were previously extrapolated from clear cell renal cell carcinoma with mixed results. More recently, some phase 2 trials focused on PRCC have been promising. Tyrosine kinase inhibitor (TKI) monotherapy is considered the standard of care, and combination strategies with TKIs and immune checkpoint inhibitors are emerging. Genetic profiling and large-scale clinical trials are needed to inform targeted treatment of PRCC.
Collapse
Affiliation(s)
- Patrick L Sweeney
- Deming Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Albert Jang
- Deming Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Shams K Halat
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Sumanta K Pal
- City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Pedro C Barata
- Deming Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA; Tulane Cancer Center, New Orleans, LA, USA; University Hospitals Seidman Cancer Center, Case Comprehensive Cancer Center, Cleveland, Ohio, USA.
| |
Collapse
|
31
|
Izarn F, Allignet B, Gille R, Boyle H, Neidhardt EM, Négrier S, Fléchon A. Real world data of diagnosis, survival, and treatment outcomes in patients with metastatic non clear cell renal cell carcinoma. Clin Genitourin Cancer 2022; 21:e35-e43. [PMID: 36272959 DOI: 10.1016/j.clgc.2022.09.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 09/19/2022] [Accepted: 09/20/2022] [Indexed: 11/03/2022]
Abstract
INTRODUCTION Metastatic non clear cell renal cell carcinoma (nccRCC) is an heterogenous group, usually excluded from phase 3 trials. We report real life data of prognosis and systemic management of those patients. METHODS We retrospectively included 102 metastatic nccRCC patients (unspecified papillary, n = 10; type 1 and 2 papillary n = 10 and n = 32; translocation RCC, n = 9; chromophobe, n = 14; collecting duct, n = 14) treated between 2006 and 2020. Objective response rate (ORR), progression-free survival (PFS) and overall survival (OS) were evaluated. RESULTS Among patients who underwent pathological review, 40.8% presented a complete histological discordance. First line treatments were mainly tyrosine kinase inhibitor (60.8%), combination including immunotherapy (7.8%) or combination of chemotherapy (13.7%). Median ORR ranged from 0% in unspecified papillary RCC to 42.9% in type 1 papillary RCC. Median PFS ranged from 2.9 months in collecting duct carcinoma to 10.9 months in type 1 papillary RCC. Median OS ranged from 6.8 months in collecting duct carcinoma to 29.1 months in MiT family translocation RCC. Thirty (29.4%) patients were included in a treatment trial during their treatment course. CONCLUSION Metastatic nccRCC patients have variable prognosis due to heterogeneity of histological subtypes. Their diagnosis and access to therapeutic innovation remain suboptimal. Dedicated prospective trials are needed.
Collapse
|
32
|
Maughan BL. Start of a New Era: Management of Non-Clear Cell Renal Cell Carcinoma in 2022. Curr Oncol Rep 2022; 24:1201-1208. [PMID: 35438388 PMCID: PMC9468090 DOI: 10.1007/s11912-022-01269-1] [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] [Accepted: 03/08/2022] [Indexed: 11/28/2022]
Abstract
PURPOSE OF REVIEW Historically, kidney cancer was diagnosed as either clear cell renal carcinoma (ccRCC) or non-clear cell renal carcinoma (nccRCC). With further research into the pathophysiology of nccRCC, multiple distinct subtypes have emerged creating distinct diagnosis, such as papillary renal cell carcinoma (PRCC), chromophobe renal cell carcinoma (crRCC), or unclassified carcinoma (cRCC). Many other kidney cancer subtypes are now included in the WHO classification system. RECENT FINDINGS The prognosis for each of the more frequently diagnosed types is discussed here along with treatment recommendations. The available clinical trial results and salient retrospective studies of each subtype are reviewed here to guide clinicians on the optimal treatment selection for patients with these rare histologic types or RCC. Many nccRCC types are now recognized and each has unique molecular drivers which are different than ccRCC. The optimal treatment strategy is different for each subtype. The prognosis also differs based on the histology.
Collapse
Affiliation(s)
- Benjamin L Maughan
- Huntsman Cancer Institute, University of Utah, 2000 Circle of Hope Drive, Room HCI S 5617, Salt Lake City, UT, 84112, USA.
| |
Collapse
|
33
|
Major heritable renal cell carcinoma syndromes: novel treatment options and future perspectives. Curr Opin Urol 2022; 32:488-494. [PMID: 35855559 DOI: 10.1097/mou.0000000000001030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
PURPOSE OF REVIEW To provide an overview of diagnosis, genetic abnormalities, clinical signs and treatment options for the major heritable renal cell carcinoma (RCC) syndromes. RECENT FINDINGS RCC in major hereditary syndromes are disorders which are typically autosomal dominant. They predispose patients to early onset of RCC and may exhibit other extrarenal manifestations. Early recognition of these diseases allows correct screening at appropriate ages as well as early detection of RCC. Moreover, expedient identification may optimize the management of extra renal manifestations as well as allow for genetic testing and screening of at-risk relatives. SUMMARY The risk of RCC in these major heritable syndromes is higher than sporadic disease. They occur at earlier age groups and can be multifocal or bilateral. Tumours are observed until at least 3 cm before any intervention, while nephron sparing surgery is widely considered as the treatment of choice except for hereditary leiomyomatosis with renal cell cancer, of which radical nephrectomy is treatment of choice. Intervention should be timeous as there is a highly reported incidence of early metastasis. Molecular therapies have been used in the setting of patients with metastasis, some of which show favourable outcomes.
Collapse
|
34
|
Lindner AK, Tulchiner G, Seeber A, Siska PJ, Thurnher M, Pichler R. Targeting strategies in the treatment of fumarate hydratase deficient renal cell carcinoma. Front Oncol 2022; 12:906014. [PMID: 35912170 PMCID: PMC9337267 DOI: 10.3389/fonc.2022.906014] [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: 03/28/2022] [Accepted: 06/29/2022] [Indexed: 11/26/2022] Open
Abstract
Fumarate hydratase (FH) - deficient renal cell carcinoma (FHdRCC) is a rare aggressive subtype of RCC caused by a germline or sporadic loss-of-function mutation in the FH gene. Here, we summarize how FH deficiency results in the accumulation of fumarate, which in turn leads to activation of hypoxia-inducible factor (HIF) through inhibition of prolyl hydroxylases. HIF promotes tumorigenesis by orchestrating a metabolic switch to glycolysis even under normoxia, a phenomenon well-known as the Warburg effect. HIF activates the transcription of many genes, including vascular endothelial growth factor (VEGF). Crosstalk between HIF and epidermal growth factor receptor (EGFR) has also been described as a tumor-promoting mechanism. In this review we discuss therapeutic options for FHdRCC with a focus on anti-angiogenesis and EGFR-blockade. We also address potential targets that arise within the metabolic escape routes taken by FH-deficient cells for cell growth and survival.
Collapse
Affiliation(s)
- Andrea Katharina Lindner
- Department of Urology, Comprehensive Cancer Center Innsbruck, Medical University of Innsbruck, Innsbruck, Austria
| | - Gennadi Tulchiner
- Department of Urology, Comprehensive Cancer Center Innsbruck, Medical University of Innsbruck, Innsbruck, Austria
| | - Andreas Seeber
- Department of Haematology and Oncology, Comprehensive Cancer Center Innsbruck, Medical University of Innsbruck, Innsbruck, Austria
| | - Peter J. Siska
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| | - Martin Thurnher
- Immunotherapy Unit, Department of Urology, Medical University of Innsbruck, Innsbruck, Austria
| | - Renate Pichler
- Department of Urology, Comprehensive Cancer Center Innsbruck, Medical University of Innsbruck, Innsbruck, Austria
- *Correspondence: Renate Pichler,
| |
Collapse
|
35
|
Abern MR, Dudinec JV, Inman BA. Genomic Sequencing Should Not be Part of the Standard of Care for Most Urologic Cancers. Eur Urol Focus 2022; 8:639-640. [DOI: 10.1016/j.euf.2022.04.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 04/25/2022] [Indexed: 11/04/2022]
|
36
|
Feng Z, Curti BD, Quinn DI, Strother JM, Chen Z, Agnor R, Beer TM, Ryan CW. A Phase II, Single-Arm Trial of Sunitinib and Erlotinib in Advanced Renal Cell Carcinoma. Clin Genitourin Cancer 2022; 20:415-422. [DOI: 10.1016/j.clgc.2022.04.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 04/27/2022] [Accepted: 04/29/2022] [Indexed: 11/25/2022]
|
37
|
Xu Y, Huang Y, Zhang J. Re: Phase II Trial of Cabozantinib Plus Nivolumab in Patients with Non-clear-cell Renal Cell Carcinoma and Genomic Correlates. Eur Urol 2022; 82:331-332. [DOI: 10.1016/j.eururo.2022.04.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 04/28/2022] [Indexed: 11/04/2022]
|
38
|
Bigot C, Boudier P, Ladoire S, Barthélémy P. Advanced nccRCC: what therapeutic options in 2022? Bull Cancer 2022; 109:2S39-2S46. [DOI: 10.1016/s0007-4551(22)00237-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
39
|
Dawsey SJ, Gupta S. Hereditary Renal Cell Carcinoma. KIDNEY CANCER 2022. [DOI: 10.3233/kca-210008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND: Hereditary renal cell carcinoma (RCC) is a complex and rapidly evolving topic as there is a growing body of literature regarding inherited syndromes and mutations associated with an increased risk of RCC. OBJECTIVES: We sought to systematically review 13 hereditary syndromes associated with RCC; von Hippel-Lindau Disease associated RCC (VHLRCC), BAP-1 associated clear cell RCC (BAPccRCC), Familial non-von Hippel Lindau clear cell RCC (FccRCC), Tuberous Sclerosis Complex associated RCC (TSCRCC), Birt-Hogg-Dub e ´ Syndrome associated RCC (BHDRCC), PTEN Hamartoma Tumor Syndrome associated RCC (PHTSRCC), Microphthalmia-associated Transcription Family translocation RCC (MiTFtRCC), RCC with Chromosome 6p Amplification (TFEBRCC), Autosomal Dominant Polycystic Kidney Disease Associated RCC (ADPKDRCC), Hereditary Leiomyomatosis associated RCC (HLRCC), Succinate Dehydrogenase RCC (SDHRCC), Hereditary Papillary RCC (HPRCC), and ALK-Rearrangement RCC (ALKRCC). RESULTS: Hereditary RCC is generally associated with early age of onset, multifocal and/or bilateral lesions, and aggressive disease course. VHLRCC, BAPccRCC, FccRCC, and certain mutations resulting in SDHRCC are associated with clear cell RCC (ccRCC). HPRCC is associated with Type 1 papillary RCC. HLRCC is associated with type 2 papillary RCC. BHDRCC is associated with Chromophobe RCC. TSCRCC, PHTSRCC, MiTFtRCC, TFEBRCC, ADPKDRCC, certain SDHRCC and ALKRCC have variable histology. CONCLUSIONS: There has been tremendous advancement in our understanding of the pathophysiology of hereditary RCC. Ongoing research will refine our understanding of hereditary RCC and its therapeutic targets.
Collapse
Affiliation(s)
- Scott J. Dawsey
- Taussig Cancer Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Shilpa Gupta
- Taussig Cancer Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| |
Collapse
|
40
|
Lee CH, Voss MH, Carlo MI, Chen YB, Zucker M, Knezevic A, Lefkowitz RA, Shapnik N, Dadoun C, Reznik E, Shah NJ, Owens CN, McHugh DJ, Aggen DH, Laccetti AL, Kotecha R, Feldman DR, Motzer RJ. Phase II Trial of Cabozantinib Plus Nivolumab in Patients With Non-Clear-Cell Renal Cell Carcinoma and Genomic Correlates. J Clin Oncol 2022; 40:2333-2341. [PMID: 35298296 DOI: 10.1200/jco.21.01944] [Citation(s) in RCA: 107] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE To assess the efficacy and safety of cabozantinib plus nivolumab in a phase II trial in patients with non-clear-cell renal cell carcinoma (RCC). PATIENTS AND METHODS Patients had advanced non-clear-cell renal carcinoma who underwent 0-1 prior systemic therapies excluding prior immune checkpoint inhibitors. Patients received cabozantinib 40 mg once daily plus nivolumab 240 mg once every 2 weeks or 480 mg once every 4 weeks. Cohort 1 enrolled patients with papillary, unclassified, or translocation-associated RCC; cohort 2 enrolled patients with chromophobe RCC. The primary end point was objective response rate (ORR) by RECIST 1.1; secondary end points included progression-free survival, overall survival, and safety. Next-generation sequencing results were correlated with response. RESULTS A total of 47 patients were treated with a median follow-up of 13.1 months. Objective response rate for cohort 1 (n = 40) was 47.5% (95% CI, 31.5 to 63.9), with median progression-free survival of 12.5 months (95% CI, 6.3 to 16.4) and median overall survival of 28 months (95% CI, 16.3 to not evaluable). In cohort 2 (n = 7), no responses were observed; one patient had stable disease > 1 year. Grade 3/4 treatment-related adverse events were observed in 32% treated patients. Cabozantinib and nivolumab were discontinued because of toxicity in 13% and 17% of patients, respectively. Common mutations included NF2 and FH in cohort 1 and TP53 and PTEN in cohort 2. Objective responses were seen in 10/12 patients with either NF2 or FH mutations. CONCLUSION Cabozantinib plus nivolumab showed promising efficacy in most non-clear-cell RCC variants tested in this trial, particularly those with prominent papillary features, whereas treatment effects were limited in chromophobe RCC. Genomic findings in non-clear-cell RCC variants warrant further study as predictors of response.
Collapse
Affiliation(s)
- Chung-Han Lee
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY.,Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Martin H Voss
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY.,Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Maria Isabel Carlo
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY.,Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Ying-Bei Chen
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Mark Zucker
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Andrea Knezevic
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Robert A Lefkowitz
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Natalie Shapnik
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Chloe Dadoun
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Ed Reznik
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Neil J Shah
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY.,Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Colette Ngozi Owens
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Deaglan Joseph McHugh
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY.,Department of Medicine, Weill Cornell Medical College, New York, NY
| | - David Henry Aggen
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY.,Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Andrew Leonard Laccetti
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY.,Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Ritesh Kotecha
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY.,Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Darren R Feldman
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY.,Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Robert J Motzer
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY.,Department of Medicine, Weill Cornell Medical College, New York, NY
| |
Collapse
|
41
|
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
|
42
|
Mar N, Kaakour D, Rezazadeh Kalebasty A. Renal Cell Carcinoma-Lessons in Diversity, Breakthroughs, and Challenges. JCO Oncol Pract 2022; 18:197-199. [PMID: 34550754 DOI: 10.1200/op.21.00446] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 07/02/2021] [Indexed: 11/20/2022] Open
Affiliation(s)
- Nataliya Mar
- Division of Hematology/Oncology, University of California Irvine, Irvine, CA
| | - Dalia Kaakour
- Department of Medicine, University of California Irvine, Irvine, CA
| | | |
Collapse
|
43
|
Brown JR, Calaway A, Castle E, Garcia J, Barata PC. Systematic Review of Treatment of Metastatic Non-Clear Cell Renal Cell Carcinoma. KIDNEY CANCER 2022. [DOI: 10.3233/kca-210005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Background: Metastatic and unresectable non-clear cell renal cell carcinoma comprises more than a quarter of kidney cancers but does not have standardized treatment. Non-clear renal carcinoma consists of a variety of diverse histologic subtypes, including papillary, chromophobe, collecting duct, translocation, and medullary histologies, many of which carry a poor prognosis. Many prospective clinical trials exclude these kidney cancers, and for most clinical trials of non-clear cell renal cell carcinoma, only a small number of patients are enrolled. Objective: To perform a systematic review of recently published and currently enrolling prospective clinical trials for advanced non-clear cell renal cell carcinoma. Methods: A systematic search of Pubmed and MEDLINE (Ovid) was conducted as per PRISMA guidelines to identify recent prospective clinical trials in non-clear cell renal cell carcinoma. To ensure a thorough search, terms not only included non-clear cell renal carcinoma but also molecular subtypes. A review of currently enrolling clinical trials was conducted on Clinicaltrials.gov and the EU Clinical Trials Register as well. Results: A total of 33 prospective clinical trials with published results and 10 currently enrolling clinicals trials were identified. About half (48.5%) of these studies were reported in 2020 or 2021, and 36.4% were in the first-line setting. Treatments investigated in these trials included mTOR inhibitors, VEGF- and MET-targeted tyrosine kinase inhibitors, immune checkpoint inhibitors, and combinatorial strategies. Outcomes from these data revealed a wide range of response rate and progression free survival, favoring TKIs and immune checkpoint inhibitors -based combination regimens. Conclusions: Novel targeted therapies and immunotherapies have changed the landscape of treatment for advanced non-clear cell renal cell carcinoma. Combination regimens may provide even further clinical benefit and warrant further investigation in larger, randomized prospective clinical trials.
Collapse
Affiliation(s)
- Jason R. Brown
- Division of Solid Tumor Oncology, UH Cleveland Medical Center, Cleveland, OH, USA
| | - Adam Calaway
- Department of Urology, UH Cleveland Medical Center, Cleveland, OH, USA
| | - Erik Castle
- Department of Urology, Tulane University Medical School, New Orleans, LA, USA
| | - Jorge Garcia
- Division of Solid Tumor Oncology, UH Cleveland Medical Center, Cleveland, OH, USA
| | - Pedro C. Barata
- Deming Department of Medicine, Section of Hematology/Medical Oncology, Tulane University Medical School, New Orleans, LA, USA
| |
Collapse
|
44
|
Medina López RA, Rivero Belenchon I, Mazuecos-Quirós J, Congregado-Ruíz CB, Couñago F. Update on the treatment of metastatic renal cell carcinoma. World J Clin Oncol 2022; 13:1-8. [PMID: 35116228 PMCID: PMC8790301 DOI: 10.5306/wjco.v13.i1.1] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Revised: 10/05/2021] [Accepted: 01/10/2022] [Indexed: 02/06/2023] Open
Abstract
Metastatic renal cell cancer (mRCC) management has undergone a paradigm shift in recent decades. The first revolution came with the emergence of vascular endothelial growth factor inhibitors; there was a second wave with the unprecedented success of checkpoint inhibitors, and then the latest approach, which is becoming the new care standard in mRCC, of combining these two strategies in different ways. Updated results of Checkmate-214 after 42 mo of follow-up were consistent with previously published results showing the superiority of nivolumab/ipilimumab over sunitinib in progression free survival (PFS), overall survival (OS), and objective response rate (ORR) in intermediate and high-risk patients. However, several studies presented at the American Society of Clinical Oncology 2020 suggested that the best place, and so far, the only one for nivolumab/ipilimumab is the frontline setting. The update on Keynote-426 after 23 mo of follow-up showed no superiority of pembroli-zumab/axitinib over sunitinib in favorable-risk mRCC, suggesting that it should no longer be the first line of choice in low-risk patients. Finally, the phase III Checkmate 9ER trial results revealed the superiority of nivolumab/cabozantinib vs sunitinib in PFS, OS, and ORR, providing a new first-line option among all International Metastatic RCC Database Consortium risk patients. Some phase II clinical trials also presented this year showed promising results with new combination therapies such as nivolumab/sitravatinib, cabozantinib/atezolizumab, and lenvatinib/pembrolizumab, providing promising grounds upon which to start phase III studies. In addition, other works are using novel therapeutic agents with different mechanisms of action, including telaglenastat (a glutaminase inhibitor), entinostat [an inhibitor of histone deacetylases (HDACs)], and olaparib and talazoparib, poly(ADP-ribose) polymerase inhibitors widely used in other tumors. However, some questions regarding mRCC management still need to be addressed, such as head-to-head comparisons between the current options, treatment sequencing, non-clear cell mRCC, and the role of biomarkers to ascertain the best treatment choice.
Collapse
Affiliation(s)
- Rafael Antonio Medina López
- Department of Urology and Nephrology, Virgen del Rocío University Hospital, Biomedical Institute of Seville/CSIC/University of Seville, Sevilla 41013, Spain
| | - Ines Rivero Belenchon
- Department of Urology and Nephrology, Virgen del Rocío University Hospital, Seville 41005, Spain
| | - Javier Mazuecos-Quirós
- Department of Urology and Nephrology, Virgen del Rocío University Hospital, Biomedical Institute of Seville/CSIC/University of Seville, Sevilla 41013, Spain
| | - Carmen Belén Congregado-Ruíz
- Department of Urology and Nephrology, Virgen del Rocío University Hospital, Biomedical Institute of Seville/CSIC/University of Seville, Sevilla 41013, Spain
| | - Felipe Couñago
- Radiation Oncology, Hospital Universitario Quirónsalud, Hospital La Luz, Universidad Europea de Madrid, Madrid 28223, Spain
| |
Collapse
|
45
|
Motzer RJ, Jonasch E, Agarwal N, Alva A, Baine M, Beckermann K, Carlo MI, Choueiri TK, Costello BA, Derweesh IH, Desai A, Ged Y, George S, Gore JL, Haas N, Hancock SL, Kapur P, Kyriakopoulos C, Lam ET, Lara PN, Lau C, Lewis B, Madoff DC, Manley B, Michaelson MD, Mortazavi A, Nandagopal L, Plimack ER, Ponsky L, Ramalingam S, Shuch B, Smith ZL, Sosman J, Dwyer MA, Gurski LA, Motter A. Kidney Cancer, Version 3.2022, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw 2022; 20:71-90. [PMID: 34991070 DOI: 10.6004/jnccn.2022.0001] [Citation(s) in RCA: 365] [Impact Index Per Article: 121.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The NCCN Guidelines for Kidney Cancer focus on the screening, diagnosis, staging, treatment, and management of renal cell carcinoma (RCC). Patients with relapsed or stage IV RCC typically undergo surgery and/or receive systemic therapy. Tumor histology and risk stratification of patients is important in therapy selection. The NCCN Guidelines for Kidney Cancer stratify treatment recommendations by histology; recommendations for first-line treatment of ccRCC are also stratified by risk group. To further guide management of advanced RCC, the NCCN Kidney Cancer Panel has categorized all systemic kidney cancer therapy regimens as "Preferred," "Other Recommended Regimens," or "Useful in Certain Circumstances." This categorization provides guidance on treatment selection by considering the efficacy, safety, evidence, and other factors that play a role in treatment selection. These factors include pre-existing comorbidities, nature of the disease, and in some cases consideration of access to agents. This article summarizes surgical and systemic therapy recommendations for patients with relapsed or stage IV RCC.
Collapse
Affiliation(s)
| | - Eric Jonasch
- The University of Texas MD Anderson Cancer Center
| | | | - Ajjai Alva
- University of Michigan Rogel Cancer Center
| | | | | | | | | | | | | | - Arpita Desai
- UCSF Helen Diller Family Comprehensive Cancer Center
| | - Yasser Ged
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins
| | | | - John L Gore
- Fred Hutchinson Cancer Research Center/Seattle Cancer Care Alliance
| | - Naomi Haas
- Abramson Cancer Center at the University of Pennsylvania
| | | | - Payal Kapur
- UT Southwestern Simmons Comprehensive Cancer Center
| | | | | | | | | | | | | | | | | | - Amir Mortazavi
- The Ohio State University Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute
| | | | | | - Lee Ponsky
- Case Comprehensive Cancer Center/ University Hospitals Seidman Cancer Center and Cleveland Clinic Taussig Cancer Institute
| | | | | | - Zachary L Smith
- Siteman Cancer Center at Barnes- Jewish Hospital and Washington University School of Medicine
| | - Jeffrey Sosman
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University
| | | | | | | |
Collapse
|
46
|
Truong H, Carlo MI. Hereditary Cancer and Genetics in Renal Cell Carcinoma. Urol Oncol 2022. [DOI: 10.1007/978-3-030-89891-5_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
47
|
Tran J, Ornstein MC. Clinical Review on the Management of Metastatic Renal Cell Carcinoma. JCO Oncol Pract 2021; 18:187-196. [PMID: 34529499 DOI: 10.1200/op.21.00419] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Renal cell carcinomas vary considerably in their tumor biology and disease course, which is reflected in the range of treatment paradigms in localized and metastatic renal cell carcinoma (mRCC). Active surveillance remains an important component of all renal cell carcinoma management. In mRCC, the rapid evolution from cytokine-based therapy to targeted therapy to immunotherapy with checkpoint blockade has revolutionized the field and drastically altered treatment outcomes. More recently, combination therapies have become a standard of care for most patients with mRCC. In this review, we highlight recent critical data that led to changes in treatment paradigms and provide a practical framework for the management of patients with mRCC.
Collapse
Affiliation(s)
- Jennifer Tran
- Department of Internal Medicine, Cleveland Clinic, Cleveland, OH
| | - Moshe C Ornstein
- Department of Hematology & Medical Oncology, Cleveland Clinic Taussig Cancer Institute, Cleveland, OH
| |
Collapse
|
48
|
Rupp N, Moch H. [FH-deficient renal cell carcinoma expands the spectrum of renal papillary tumors]. DER PATHOLOGE 2021; 42:560-564. [PMID: 34448900 PMCID: PMC8536582 DOI: 10.1007/s00292-021-00977-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 07/09/2021] [Indexed: 11/30/2022]
Abstract
Fumarate hydratase (FH)-deficient renal cell carcinoma (RCC) is a distinct entity, which shows a biallelic inactivation of the FH gene that consequently leads to FH protein expression and function loss, respectively. This alteration leads to an accumulation of the oncometabolite fumarate in the citrate cycle and various disorders of the cell balance and DNA processing. FH-deficient RCC often shows a morphologically overlapping spectrum with papillary renal cell carcinoma (type 2), whereby a typical mixture of growth patterns including tubulo-cystic, cribriform, and/or solid differentiation can be observed. A characteristic but non-specific morphological feature is prominent eosinophilic, virus-inclusion body-like nucleoli with perinucleolar halos. Tumoral immunohistochemical loss of FH expression supports the diagnosis but may be preserved in rare cases. Most FH-deficient RCCs show very aggressive biological behavior and are often metastasized at the time of diagnosis. The initial description encompassed RCC in association with the hereditary leiomyomatosis and renal cell carcinoma (HLRCC) syndrome, which also includes cutaneous and uterine leiomyomas. However, current data also show an increasing proportion of sporadic cases, so that a distinction (hereditary vs. sporadic) seems appropriate. So far, few but promising data on effective systemic therapeutic options have been reported. In summary, precise diagnosis is of great importance due to the frequent aggressive biological behavior, potential need to deviate from the therapeutic standard, and the possible indicator of a hereditary disease.
Collapse
Affiliation(s)
- N Rupp
- Institut für Pathologie und Molekularpathologie, Universitätsspital Zürich, Schmelzbergstr. 12, 8091, Zürich, Schweiz. .,Universität Zürich, Zürich, Schweiz.
| | - H Moch
- Institut für Pathologie und Molekularpathologie, Universitätsspital Zürich, Schmelzbergstr. 12, 8091, Zürich, Schweiz.,Universität Zürich, Zürich, Schweiz
| |
Collapse
|
49
|
Stadler ZK, Maio A, Chakravarty D, Kemel Y, Sheehan M, Salo-Mullen E, Tkachuk K, Fong CJ, Nguyen B, Erakky A, Cadoo K, Liu Y, Carlo MI, Latham A, Zhang H, Kundra R, Smith S, Galle J, Aghajanian C, Abu-Rustum N, Varghese A, O'Reilly EM, Morris M, Abida W, Walsh M, Drilon A, Jayakumaran G, Zehir A, Ladanyi M, Ceyhan-Birsoy O, Solit DB, Schultz N, Berger MF, Mandelker D, Diaz LA, Offit K, Robson ME. Therapeutic Implications of Germline Testing in Patients With Advanced Cancers. J Clin Oncol 2021; 39:2698-2709. [PMID: 34133209 PMCID: PMC8376329 DOI: 10.1200/jco.20.03661] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 04/30/2021] [Accepted: 05/06/2021] [Indexed: 12/27/2022] Open
Abstract
PURPOSE Tumor mutational profiling is increasingly performed in patients with advanced cancer. We determined the extent to which germline mutation profiling guides therapy selection in patients with advanced cancer. METHODS Patients with cancer undergoing tumor genomic profiling were prospectively consented for germline cancer predisposition gene analysis (2015-2019). In patients harboring germline likely pathogenic or pathogenic (LP/P) alterations, therapeutic actionability was classified using a precision oncology knowledge base. Patients with metastatic or recurrent cancer receiving germline genotype-directed therapy were determined. RESULTS Among 11,947 patients across > 50 malignancies, 17% (n = 2,037) harbored a germline LP/P variant. By oncology knowledge base classification, 9% (n = 1042) had an LP/P variant in a gene with therapeutic implications (4% level 1; 4% level 3B; < 1% level 4). BRCA1/2 variants accounted for 42% of therapeutically actionable findings, followed by CHEK2 (13%), ATM (12%), mismatch repair genes (11%), and PALB2 (5%). When limited to the 9,079 patients with metastatic or recurrent cancer, 8% (n = 710) harbored level 1 or 3B genetic findings and 3.2% (n = 289) received germline genotype-directed therapy. Germline genotype-directed therapy was received by 61% and 18% of metastatic cancer patients with level 1 and level 3B findings, respectively, and by 54% of BRCA1/2, 75% of mismatch repair, 43% of PALB2, 35% of RAD51C/D, 24% of BRIP1, and 19% of ATM carriers. Of BRCA1/2 patients receiving a poly(ADP-ribose) polymerase inhibitor, 45% (84 of 188) had tumors other than breast or ovarian cancer, wherein the drug, at time of delivery, was delivered in an investigational setting. CONCLUSION In a pan-cancer analysis, 8% of patients with advanced cancer harbored a germline variant with therapeutic actionability with 40% of these patients receiving germline genotype-directed treatment. Germline sequence analysis is additive to tumor sequence analysis for therapy selection and should be considered for all patients with advanced cancer.
Collapse
Affiliation(s)
- Zsofia K. Stadler
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Anna Maio
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Debyani Chakravarty
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Yelena Kemel
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Margaret Sheehan
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Erin Salo-Mullen
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Kaitlyn Tkachuk
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Christopher J. Fong
- Computational Oncology, Department of Epidemiology and Statistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Bastien Nguyen
- Computational Oncology, Department of Epidemiology and Statistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Amanda Erakky
- David M. Rubinstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Karen Cadoo
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Ying Liu
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Maria I. Carlo
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Alicia Latham
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Hongxin Zhang
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Ritika Kundra
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Shaleigh Smith
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Jesse Galle
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Carol Aghajanian
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Nadeem Abu-Rustum
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Anna Varghese
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Eileen M. O'Reilly
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- David M. Rubinstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Michael Morris
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Wassim Abida
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Michael Walsh
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Alexander Drilon
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Gowtham Jayakumaran
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Ahmet Zehir
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Marc Ladanyi
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Ozge Ceyhan-Birsoy
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - David B. Solit
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Nikolaus Schultz
- Computational Oncology, Department of Epidemiology and Statistics, Memorial Sloan Kettering Cancer Center, New York, NY
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Michael F. Berger
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Diana Mandelker
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Luis A. Diaz
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Kenneth Offit
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Mark E. Robson
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
50
|
Sepe P, Ottini A, Pircher CC, Franza A, Claps M, Guadalupi V, Verzoni E, Procopio G. Characteristics and Treatment Challenges of Non-Clear Cell Renal Cell Carcinoma. Cancers (Basel) 2021; 13:3807. [PMID: 34359706 PMCID: PMC8345088 DOI: 10.3390/cancers13153807] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 07/15/2021] [Accepted: 07/26/2021] [Indexed: 01/05/2023] Open
Abstract
Non-clear cell renal cell carcinomas (RCC) comprise several rare and poorly described diseases, often characterized by bad prognosis and with no standard treatments available. The gap in their clinical management is linked to the poor molecular characterization in handling the treatment of non clear-cell RCC with untailored therapies. Due to their rarity, non-clear RCC are in fact under-represented in prospective randomized trials. Thus, treatment choices are based on extrapolating results from clear cell RCC trials, retrospective data, or case reports. Over the last two decades, various options have been considered as the mainstay for the treatment of metastatic RCC (mRCC), including angiogenesis inhibitors, vascular endothelial growth factor receptor inhibitors, other tyrosine kinase inhibitors (TKIs), as well as MET inhibitors and mammalian targeting of rapamycin (mTOR) inhibitors. More recently, the therapeutic armamentarium has been enriched with immunotherapy, alone or in combination with targeted agents that have been shown to significantly improve outcomes of mRCC patients, if compared to TKI single-agent. It has been widely proven that non-clear cell RCC is a morphologically and clinically distinct entity from its clear cell counterpart but more knowledge about its biology is certainly needed. Histology-specific collaborative trials are in fact now emerging to investigate different treatments for non-clear cell RCC. This review summarizes pathogenetic mechanisms of non-clear cell RCC, the evolution of treatment paradigms over the last few decades, with a focus on immunotherapy-based trials, and future potential treatment options.
Collapse
Affiliation(s)
- Pierangela Sepe
- Department of Medical Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, 20133 Milan, Italy; (A.O.); (C.C.P.); (A.F.); (M.C.); (V.G.); (E.V.); (G.P.)
| | | | | | | | | | | | | | | |
Collapse
|