1
|
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
|
2
|
Barata P, Tangen C, Plets M, Thompson IM, Narayan V, George DJ, Heng DYC, Shuch B, Stein M, Gulati S, Tretiakova M, Tripathi A, Bjarnason GA, Humphrey P, Adeniran A, Vaishampayan U, Alva A, Zhang T, Cole S, Lara PN, Lerner SP, Balzer-Haas N, Pal SK. Final Overall Survival Analysis of S1500: A Randomized, Phase II Study Comparing Sunitinib With Cabozantinib, Crizotinib, and Savolitinib in Advanced Papillary Renal Cell Carcinoma. J Clin Oncol 2024; 42:3911-3916. [PMID: 39255440 PMCID: PMC11575905 DOI: 10.1200/jco.24.00767] [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: 04/09/2024] [Revised: 06/11/2024] [Accepted: 07/09/2024] [Indexed: 09/12/2024] Open
Abstract
Clinical trials frequently include multiple end points that mature at different times. The initial report, typically based on the primary end point, may be published when key planned co-primary or secondary analyses are not yet available. Clinical Trial Updates provide an opportunity to disseminate additional results from studies, published in JCO or elsewhere, for which the primary end point has already been reported.Mesenchymal-epithelial transition (MET) signaling pathway plays a role in the pathogenesis of selected patients with papillary renal cell carcinoma (PRCC). In the phase II PAPMET trial (ClinicalTrials.gov identifier: NCT02761057), cabozantinib significantly prolonged progression-free survival and improved objective response rate compared with sunitinib in patients with advanced PRCC. Here, we present the final overall survival (OS) analysis. In this multicenter, randomized phase II, open-label trial, 147 patients with advanced PRCC who have received up to one previous therapy (excluding vascular endothelial growth factor-directed agents) were assigned to sunitinib, cabozantinib, crizotinib, or savolitinib. Ultimately, savolitinib and crizotinib arms were closed because of futility. With a median follow-up of 17.5 months, the median OS was 21.5 months (95% CI, 12.0 to 28.1) with cabozantinib and 17.3 months (95% CI, 12.8 to 21.8) with sunitinib (hazard ratio, 0.83; 95% CI, 0.51 to 1.36; P = .46). The OS landmark estimates for cabozantinib and sunitinib were 50% versus 39% at 24 months and 32% versus 28% at 36 months. In conclusion, we observed no significant difference in OS across treatment arms. Although cabozantinib represents a well-supported option for advanced PRCC, the lack of survival benefit underscores the need to develop novel therapies for this disease.
Collapse
Affiliation(s)
- Pedro Barata
- University Hospitals Seidman Cancer Center, Cleveland, OH
| | - Catherine Tangen
- SWOG Statistics and Data Management Center, Seattle, WA
- Fred Hutchinson Cancer Center, Seattle, WA
| | - Melissa Plets
- SWOG Statistics and Data Management Center, Seattle, WA
- Fred Hutchinson Cancer Center, Seattle, WA
| | - Ian M Thompson
- CHRISTUS Santa Rosa Medical Center Hospital, Houston, TX
| | - Vivek Narayan
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| | | | | | - Brian Shuch
- Institute of Urologic Oncology, UCLA, Los Angeles, CA
| | | | - Shuchi Gulati
- UC Davis Comprehensive Cancer Center, Sacramento, CA
| | | | | | | | | | | | | | | | - Tian Zhang
- University of Texas Southwestern Medical Center, Dallas, TX
| | - Scott Cole
- Oklahoma Cancer Specialists and Research Institute, Tulsa, OK
| | - Primo N Lara
- UC Davis Comprehensive Cancer Center, Sacramento, CA
| | | | - Naomi Balzer-Haas
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| | - Sumanta K Pal
- City of Hope Comprehensive Cancer Center, Duarte, CA
| |
Collapse
|
3
|
Ishihara H, Nemoto Y, Mizoguchi S, Nishimura K, Ikeda T, Fukuda H, Yoshida K, Shimmura H, Hashimoto Y, Iizuka J, Kondo T, Takagi T. Changes in outcome of patients with advanced non-clear cell renal cell carcinoma from the tyrosine kinase inhibitor era to the immuno-oncology era. Int J Clin Oncol 2024; 29:1730-1739. [PMID: 39143429 DOI: 10.1007/s10147-024-02606-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: 05/18/2024] [Accepted: 08/04/2024] [Indexed: 08/16/2024]
Abstract
BACKGROUND The therapeutic benefit of immuno-oncology (IO) therapy for patients with advanced non-clear-cell renal cell carcinoma (nccRCC) remains unclear. PATIENTS AND METHODS We reviewed clinical data from 93 patients with advanced nccRCC who received first-line systemic therapy including IO combination therapy and tyrosine kinase inhibitor (TKI) monotherapy at our affiliated institutions. Patients were divided based on the period when the treatment was implemented as the standard of care into the IO and TKI eras. Survival and tumor response outcomes were compared between the IO and TKI eras. RESULTS Of the 93 patients, 50 (54%) and 43 (46%) were categorized as IO era and TKI era groups, respectively. Progression-free survival (PFS) and overall survival (OS) were significantly longer in the IO era than in the TKI era (median PFS: 8.97 vs. 4.96 months, p = 0.0152; median OS: 38.4 vs. 13.5 months, p = 0.0001). After the adjustment using other covariates, the treatment era was an independent factor for PFS (hazard ratio: 0.59, p = 0.0235) and OS (hazard ratio: 0.27, p < 0.0001). Objective response and disease control rates was not significantly different between the treatment eras (26% vs. 16.3%, p = 0.268; 62% vs. 62.8%, p = 0.594). CONCLUSION The implementation of IO therapy was significantly associated with longer survival in the nccRCC population. Further studies are needed to establish a more effective treatment strategy in this population using multiple regimens of IO combination therapy.
Collapse
Affiliation(s)
- Hiroki Ishihara
- Department of Urology, Saiseikai Kawaguchi General Hospital, 5-11-5 Nishikawaguchi, Kawaguchi, Saitama, Japan.
- Department of Urology, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, Japan.
| | - Yuki Nemoto
- Department of Urology, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, Japan
- Department of Urology, Jyoban Hospital, Uenodai 57, Joban Kamiyunagayamachi, Iwaki, Fukushima, Japan
| | - Shinsuke Mizoguchi
- Department of Urology, Saiseikai Kazo Hospital, 1680 Kamitakayanagi, Kazo, Saitama, Japan
| | - Koichi Nishimura
- Department of Urology, Tokyo Women's Medical University Adachi Medical Center, 4-33-1 Kouhoku, Adachi-ku, Tokyo, Japan
| | - Takashi Ikeda
- Department of Urology, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, Japan
| | - Hironori Fukuda
- Department of Urology, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, Japan
| | - Kazuhiko Yoshida
- Department of Urology, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, Japan
| | - Hiroaki Shimmura
- Department of Urology, Jyoban Hospital, Uenodai 57, Joban Kamiyunagayamachi, Iwaki, Fukushima, Japan
| | - Yasunobu Hashimoto
- Department of Urology, Saiseikai Kawaguchi General Hospital, 5-11-5 Nishikawaguchi, Kawaguchi, Saitama, Japan
| | - Junpei Iizuka
- Department of Urology, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, Japan
| | - Tsunenori Kondo
- Department of Urology, Tokyo Women's Medical University Adachi Medical Center, 4-33-1 Kouhoku, Adachi-ku, Tokyo, Japan
| | - Toshio Takagi
- Department of Urology, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, Japan
| |
Collapse
|
4
|
Tripathi A, Tangen CM, Plets M, Li X, Tretiakova M, Humphrey PA, Adeniran A, Barata PC, Gulati S, Bergerot CD, Pruthi DK, Thompson IM, Lara PN, Lerner SP, Pal SK, Shuch BM. Pathological concordance rate and outcomes by subtype in advanced papillary renal cell carcinoma. BJU Int 2024; 134:596-601. [PMID: 39014969 DOI: 10.1111/bju.16403] [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] [Indexed: 07/18/2024]
Abstract
OBJECTIVE To evaluate the clinical significance of subtyping (type 1 vs 2) of papillary renal cell carcinoma (PRCC) in patients treated with targeted therapy, as well as the concordance, sensitivity and positive predictive value (PPV) of local review pathology review. METHODS Patients with advanced refractory PRCC were randomised to receive sunitinib or cabozantinib, crizotinib or savolitinib, stratified by PRCC subtype (type 1, type 2, or not otherwise specified [NOS]/mixed) by local review. Central review was retrospectively conducted by three expert genitourinary pathologists who independently reviewed cases. The sensitivity and PPV of local review were estimated and outcomes [objective response rate (ORR), progression-free survival (PFS)] were summarised for treatment groups stratified by subtypes by central review. RESULTS Amongst the 147 patients reviewed, the prevalence of individual subtypes varied by local or central review (type 1: 17.7% vs 29.3%; type 2: 53.1% vs 45.6%; NOS/mixed: 29.3% vs 25.2%), respectively. Individual cases were frequently reclassified and local pathology review demonstrated low sensitivity (type 1: 48%, 95% confidence interval [CI] 33, 65; type 2: 67%, 95% CI 55, 78; NOS/mixed: 43%, 95% CI 27, 61). The PPVs of local review were 80%, 57.7% and 37% for type 1, 2 and NOS/mixed, respectively. Compared to sunitinib, cabozantinib demonstrated improved PFS for both type 1 and type 2 PRCC subgroups (7.4 vs 9.0 and 2.9 vs 5.6 months, respectfully) as well as higher ORR. CONCLUSIONS The PRCC subtype assignment did not identify a subset of patients with greater clinical benefit from cabozantinib, with significant discordance between local and central review. Our findings confirm the limited clinical value of pathological subtyping of metastatic PRCC, in line with the recent World Health Organisation 2022 guidelines. PATIENT SUMMARY In this study, categorising papillary renal cell carcinoma into type 1 or 2 subtypes showed limited concordance between central and local pathological review and did not enrich for patients more likely to benefit from cabozantinib in the S1500 PAPMET trial.
Collapse
Affiliation(s)
| | | | - Melissa Plets
- SWOG Statistics and Data Management Center, Seattle, WA, USA
| | - Xiaochen Li
- Department of Biostatistics, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | | | | | | | - Pedro C Barata
- University Hospitals Seidman Cancer Center, Cleveland, OH, USA
| | - Shuchi Gulati
- University of California Davis Comprehensive Cancer Center, Sacramento, CA, USA
| | | | | | - Ian M Thompson
- Children's Hospital of San Antonio, San Antonio, TX, USA
| | - Primo N Lara
- University of California Davis Comprehensive Cancer Center, Sacramento, CA, USA
| | - Seth P Lerner
- Baylor College of Medicine, Dan L Duncan Cancer Center, Houston, TX, USA
| | - Sumanta K Pal
- City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Brian M Shuch
- UCLA Institute of Urologic Oncology, Los Angeles, CA, USA
| |
Collapse
|
5
|
Kotecha RR, Doshi SD, Knezevic A, Chaim J, Chen Y, Jacobi R, Zucker M, Reznik E, McHugh D, Shah NJ, Feld E, Aggen DH, Rafelson W, Xiao H, Carlo MI, Feldman DR, Lee CH, Motzer RJ, Voss MH. A Phase 2 Trial of Talazoparib and Avelumab in Genomically Defined Metastatic Kidney Cancer. Eur Urol Oncol 2024; 7:804-811. [PMID: 37945488 PMCID: PMC11074239 DOI: 10.1016/j.euo.2023.10.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 08/07/2023] [Accepted: 10/20/2023] [Indexed: 11/12/2023]
Abstract
BACKGROUND Although different kidney cancers represent a heterogeneous group of malignancies, multiple subtypes including Von Hippel-Lindau (VHL)-altered clear cell renal cell carcinoma (ccRCC), fumarate hydratase (FH)- and succinate dehydrogenase (SDH)-deficient renal cell carcinoma (RCC), and renal medullary carcinoma (RMC) are affected by genomic instability. Synthetic lethality with poly ADP-ribose polymerase inhibitors (PARPis) has been suggested in preclinical models of these subtypes, and paired PARPis with immune checkpoint blockade (ICB) may achieve additive and/or synergistic effects in patients with previously treated advanced kidney cancers. OBJECTIVE To evaluate combined PARPi + ICB in treatment-refractory metastatic kidney cancer. DESIGN, SETTING, AND PARTICIPANTS We conducted a single-center, investigator-initiated phase 2 trial in two genomically selected advanced kidney cancer cohorts: (1) VHL-altered RCC with at least one prior ICB agent and one vascular endothelial growth factor (VEGF) inhibitor, and (2) FH- or SDH-deficient RCC with at least one prior ICB agent or VEGF inhibitor and RMC with at least one prior line of chemotherapy. INTERVENTION Patients received talazoparib 1 mg daily plus avelumab 800 mg intravenously every 14 d in 28-d cycles. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS The primary endpoint was objective response rate (ORR) by Immune Response Evaluation Criteria in Solid Tumors at 4 mo, and the secondary endpoints included progression-free survival (PFS), overall survival, and safety. RESULTS AND LIMITATIONS Cohort 1 consisted of ten patients with VHL-altered ccRCC. All patients had previously received ICB. The ORR was 0/9 patients; one patient was not evaluable due to missed doses. In this cohort, seven patients achieved stable disease (SD) as the best response. The median PFS was 3.5 mo (95% confidence interval [CI] 1.0, 3.9 mo). Cohort 2 consisted of eight patients; four had FH-deficient RCC, one had SDH-deficient RCC, and three had RMC. In this cohort, six patients had previously received ICB. The ORR was 0/8 patients; two patients achieved SD as the best response and the median PFS was 1.2 mo (95% CI 0.4, 2.9 mo). The most common treatment-related adverse events of all grades were fatigue (61%), anemia (28%), nausea (22%), and headache (22%). There were seven grade 3-4 and no grade 5 events. CONCLUSIONS The first clinical study of combination PARPi and ICB therapy in advanced kidney cancer did not show clinical benefit in multiple genomically defined metastatic RCC cohorts or RMC. PATIENT SUMMARY We conducted a study to look at the effect of two medications, talazoparib and avelumab, in patients with metastatic kidney cancer who had disease progression on standard treatment. Talazoparib blocks the normal activity of molecules called poly ADP-ribose polymerase, which then prevents tumor cells from repairing themselves and growing, while avelumab helps the immune system recognize and kill cancer cells. We found that the combination of these agents was safe but not effective in specific types of kidney cancer.
Collapse
Affiliation(s)
- Ritesh R Kotecha
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA.
| | - Sahil D Doshi
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Andrea Knezevic
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Joshua Chaim
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yingbei Chen
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Rachel Jacobi
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mark Zucker
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ed Reznik
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Deaglan McHugh
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Neil J Shah
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Emily Feld
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - David H Aggen
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - William Rafelson
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Han Xiao
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Maria I Carlo
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Darren R Feldman
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Chung-Han Lee
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Robert J Motzer
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Martin H Voss
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA.
| |
Collapse
|
6
|
Ma X, Chen J, Chen S, Lan X, Wei Z, Gao H, Hou E. Immunotherapy for renal cell carcinoma: New therapeutic combinations and adverse event management strategies: A review. Medicine (Baltimore) 2024; 103:e38991. [PMID: 39058879 PMCID: PMC11272340 DOI: 10.1097/md.0000000000038991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 06/28/2024] [Indexed: 07/28/2024] Open
Abstract
Immune checkpoint inhibitor (ICI) combinations, as well as ICIs combined with tyrosine kinase inhibitors, have considerable potential for renal cell carcinoma (RCC) treatment. Newer targeted medications, gut microbiome, nanomedicines, and cyclin-dependent kinase (CDK) inhibitors demonstrate significant potential in preventing side effects and resistance associated with RCC treatment. Most patients, including those demonstrating long-term treatment effects, eventually demonstrate cancer progression. Nevertheless, recent studies have further revealed RCC pathogenesis and many acquired drug resistance mechanisms, which together have led to the identification of promising therapeutic targets. In addition to having roles in metabolism, immunogenicity, and the immune response to tumors, CDK4 and CDK6 regulate the cell cycle. Targeting CDK4 and CDK6, either separately or in combination with already approved treatments, may improve therapeutic outcomes in patients with kidney cancer. Other novel drugs, including pegylated interleukin 10, colony-stimulating factor 1 receptor inhibitors, CD40 agonists, and C-X-C receptor 4 inhibitors affect the tumor microenvironment and cancer cell metabolism. Moreover, a triple ICI combination has been noted to be efficacious. In general, compared with sunitinib as a single-drug treatment, newer ICI combinations improve overall survival in patients with RCC. Future research on the prevention of adverse events and medication resistance related to newer therapies may aid in ensuring effective treatment outcomes among patients with RCC. This article aims to summarize innovative immunotherapy drug combinations for RCC treatment and the mechanisms of action, drug resistance, and treatment of adverse events associated with these combinations.
Collapse
Affiliation(s)
- Xiaohan Ma
- Graduate School, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Jibing Chen
- Ruikang Hospital, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Sheng Chen
- Graduate School, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Xuan Lan
- Graduate School, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Zengzhao Wei
- Graduate School, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Hongjun Gao
- Ruikang Hospital, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Encun Hou
- Ruikang Hospital, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| |
Collapse
|
7
|
Naffrichoux J, Poupin P, Pouillot W, Linassier C, Rioux-Leclercq N, De Vries-Brilland M, Mourey L, Laguerre B, Oudard S, Gross-Goupil M, Mousset C, Gravis G, Rolland F, Moise L, Emambux S, Vassal C, Zanetta S, Penel N, Albiges L, Fromont G, Cancel M. PD-L1 expression and its prognostic value in metastatic papillary renal cell carcinoma: Results from a GETUG multicenter retrospective cohort. Eur J Cancer 2024; 205:114121. [PMID: 38749111 DOI: 10.1016/j.ejca.2024.114121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 05/02/2024] [Accepted: 05/09/2024] [Indexed: 06/09/2024]
Abstract
INTRODUCTION Papillary renal cell carcinoma (pRCC) is a rare and aggressive cancer with no specifically established therapeutic strategy in the metastatic setting. Combinations of tyrosine kinase and immune checkpoint inhibitors (ICI) are a promising option. We aimed to study the immune landscape of metastatic pRCC, and its interactions with angiogenesis pathways, to search for potential therapeutic targets. METHODS The expression of immune markers (PD-L1, PD-1, PD-L2, LAG-3) and angiogenic pathways (CAIX, c-MET), was analyzed by immunohistochemistry on 68 metastatic pRCC retrieved from a retrospective multicenter GETUG cohort. Our primary endpoint was to estimate the prevalence of PD-L1 expression and its prognostic impact in metastatic pRCC. Secondary endpoints included the evaluation of other immune markers (PD-1, PD-L2, and LAG-3) and their association with PD-L1. We also assessed angiogenic markers and their association with PD-L1. RESULTS Overall, 27.9 % of tumors were PD-L1 positive. PD-L2 was more frequently expressed (45.6 %), PD-1 and LAG-3 were positive in 17.6 % and 19.1 % respectively. None of these markers was correlated with PD-L1 expression. 66 % (45/68) expressed at least one immune marker, and 43 % (29/68) were "double-positive", as they expressed both immune and angiogenic markers. OS was significantly shorter for patients with PD-L1 positive pRCC. A multivariate analysis confirmed a significant association between PD-L1 expression and shorter overall survival (HR = 4.0, p = 0.01). CONCLUSION These results reinforce clinical data on the expected benefit of ICI in metastatic pRCC treatment, as PD-L1 expression is a factor of poor prognosis in this multicenter cohort.
Collapse
Affiliation(s)
| | | | | | - Claude Linassier
- Department of Medical Oncology, University Hospital, Tours, France
| | | | | | - Loïc Mourey
- Department of Medical Oncology, IUCT Oncopole, Toulouse, France
| | - Brigitte Laguerre
- Department of Medical Oncology, Eugène Marquis Cancer Center, Rennes, France
| | - Stéphane Oudard
- Department of Medical Oncology, Georges Pompidou Hospital, University Paris Cité, Paris, France
| | - Marine Gross-Goupil
- Department of Medical Oncology, Saint-André University Hospital, Bordeaux, France
| | | | - Gwenaelle Gravis
- Department of Medical Oncology, Institut Paoli-Calmettes, Marseille, France
| | - Frédéric Rolland
- Department of Medical Oncology, Institut de Cancérologie de L'Ouest, Saint Herblain, France
| | - Laura Moise
- Department of Medical Oncology, Centre François Baclesse, Caen, France
| | - Sheik Emambux
- Department of Medical Oncology, La Milétrie University Hospital, Poitiers, France
| | - Cécile Vassal
- Department of Medical Oncology, Institut de Cancérologie Lucien Neuwirth, Saint-Priest-en-Jarez, France
| | - Sylvie Zanetta
- Department of Medical Oncology, Centre Georges-François Leclerc, Dijon, France
| | - Nicolas Penel
- Lille University and Department of Medical Oncology, Centre Oscar Lambret, Lille, France
| | - Laurence Albiges
- Department of Medical Oncology, Gustave Roussy, Villejuif, France
| | - Gaëlle Fromont
- Department of Pathology, University Hospital, Tours, France; INSERM UMR 1069, N2COx, Tours University, Tours, France
| | - Mathilde Cancel
- Department of Medical Oncology, University Hospital, Tours, France; INSERM UMR 1069, N2COx, Tours University, Tours, France.
| |
Collapse
|
8
|
Salgia NJ, Zengin ZB, Pal SK, Dizman N. Renal Cell Carcinoma of Variant Histology: New Biologic Understanding Leads to Therapeutic Advances. Am Soc Clin Oncol Educ Book 2024; 44:e438642. [PMID: 38776514 PMCID: PMC11235416 DOI: 10.1200/edbk_438642] [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] [Indexed: 05/25/2024]
Abstract
Renal cell carcinoma (RCC) is one of the 10 most commonly diagnosed solid tumors. Most RCCs are histologically defined as clear cell, comprising approximately 75% of diagnoses. However, the remaining RCC cases are composed of a heterogeneous combination of diverse histopathologic subtypes, each with unique pathogeneses and clinical features. Although the therapeutic approach to both localized and metastatic RCCs has dramatically changed, first with the advent of antiangiogenic targeted therapies and more recently with the approval of immune checkpoint inhibitor (ICI)-based combinations, these advances have primarily benefited the clear cell RCC patient population. As such, there remains critical gaps in the optimization of treatment regimens for patients with non-clear cell, or variant, RCC histologies. Herein, we detail recent advances in understanding the biology of RCC with variant histology and how such findings have guided novel clinical studies investigating precision oncology approaches for these rare subtypes. Among the most common variant histology RCCs are papillary RCC, comprising approximately 15%-20% of all diagnoses. Although a histopathologically diverse subset of tumors, papillary RCC is canonically associated with amplification of the MET protooncogene; recently completed and ongoing trials have investigated MET-directed therapies for this patient population. Finally, we discuss the unique biology of RCC with sarcomatoid dedifferentiation and the recent clinical findings detailing its paradoxical sensitivity to ICIs.
Collapse
Affiliation(s)
- Nicholas J Salgia
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY
- Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY
| | - Zeynep B Zengin
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT
| | - Sumanta K Pal
- Department of Medical Oncology & Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, CA
| | - Nazli Dizman
- Department of Hematology and Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
9
|
Coffey NJ, Simon MC. Metabolic alterations in hereditary and sporadic renal cell carcinoma. Nat Rev Nephrol 2024; 20:233-250. [PMID: 38253811 PMCID: PMC11165401 DOI: 10.1038/s41581-023-00800-2] [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] [Accepted: 11/30/2023] [Indexed: 01/24/2024]
Abstract
Kidney cancer is the seventh leading cause of cancer in the world, and its incidence is on the rise. Renal cell carcinoma (RCC) is the most common form and is a heterogeneous disease comprising three major subtypes that vary in their histology, clinical course and driver mutations. These subtypes include clear cell RCC, papillary RCC and chromophobe RCC. Molecular analyses of hereditary and sporadic forms of RCC have revealed that this complex and deadly disease is characterized by metabolic pathway alterations in cancer cells that lead to deregulated oxygen and nutrient sensing, as well as impaired tricarboxylic acid cycle activity. These metabolic changes facilitate tumour growth and survival. Specifically, studies of the metabolic features of RCC have led to the discovery of oncometabolites - fumarate and succinate - that can promote tumorigenesis, moonlighting functions of enzymes, and substrate auxotrophy owing to the disruption of pathways that enable the production of arginine and cholesterol. These metabolic alterations within RCC can be exploited to identify new therapeutic targets and interventions, in combination with novel approaches that minimize the systemic toxicity of metabolic inhibitors and reduce the risk of drug resistance owing to metabolic plasticity.
Collapse
Affiliation(s)
- Nathan J Coffey
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - M Celeste Simon
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
10
|
Nova-Camacho LM, Acosta AM, Akgul M, Panizo A, Galea LA, Val-Carreres A, Talavera JA, Guerrero-Setas D, Martin-Arruti M, Ruiz I, García-Martos M, Sangoi AR. Biphasic papillary (biphasic squamoid alveolar) renal cell carcinoma: a clinicopathologic and molecular study of 17 renal cell carcinomas including 10 papillary adenomas. Virchows Arch 2024; 484:441-449. [PMID: 38388964 DOI: 10.1007/s00428-024-03768-x] [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: 12/03/2023] [Revised: 01/12/2024] [Accepted: 02/07/2024] [Indexed: 02/24/2024]
Abstract
Biphasic papillary renal cell carcinoma (synonymous with biphasic squamoid alveolar renal cell carcinoma) is considered within the spectrum of papillary renal cell carcinoma (PRCC). With < 70 reported cases of biphasic PRCC, there is limited data on the pathologic spectrum and clinical course. Seventeen biphasic PRCC cases and 10 papillary adenomas with similar biphasic morphology were assessed. The mean age of the biphasic PRCC patients was 62 years (male to female ratio of 1.8:1), from 10 partial nephrectomies, 6 radical nephrectomies, and 1 biopsy. The mean tumor size was 3.6 cm (range 1.6-8 cm), with 24% showing multifocality. Fifteen out of 17 cases were limited to the kidney (one of which was staged as pT2a but had lung metastases at diagnosis) and 2/17 cases were staged as T3a. All tumors showed typical biphasic morphology with an extent of squamoid foci widely variable from 10 to 95%. Emperipolesis was identified in 88% of cases. All biphasic PRCC tested exhibited positivity for PAX8 (16/16), keratin 7 (17/17), EMA (15/15), AMACR (17/17), and vimentin (12/12) in both large and small cells; cyclin D1 was only expressed in the large cells (16/16). The 10 papillary adenomas showed a similar immunoprofile to biphasic PRCC. NGS testing performed on 13 biphasic PRCC revealed 4 (31%) harboring MET SNVs. In 1/5 (20%) papillary adenomas, a pathogenic MET SNV was identified. Biphasic PRCC is rare with a generally similar immunoprofile to "type 1" PRCC but with notable strong positivity for cyclin D1 in the large cell component. Although most of the biphasic PRCC cases were of small size, low stage, and with an indolent behavior, one patient had metastatic disease and one patient died of the disease.
Collapse
Affiliation(s)
- Luiz M Nova-Camacho
- Department of Pathology, Donostia University Hospital, San Sebastian, Spain.
- , Gainesville, USA.
| | - Andres M Acosta
- Department of Pathology, Indiana University, Indianapolis, IN, USA
| | - Mahmut Akgul
- Department of Pathology and Laboratory Medicine, Albany Medical Center, Albany, NY, USA
| | - Angel Panizo
- Department of Pathology, University Hospital of Navarra, Pamplona, Spain
| | - Laurence A Galea
- Department of Anatomical Pathology, Sonic Healthcare, Melbourne PathologyVictoria, Australia
| | | | - Juan A Talavera
- Department of Internal Medicine, Diagnósticos da America DASA, Sao Paulo, Brazil
| | | | - Maialen Martin-Arruti
- Department of Pathology, Donostia University Hospital, San Sebastian, Spain
- Laboratory of Molecular Pathology and Therapeutic Targets, Donostia University Hospital, San Sebastian, Spain
| | - Irune Ruiz
- Department of Pathology, Donostia University Hospital, San Sebastian, Spain
- Laboratory of Molecular Pathology and Therapeutic Targets, Donostia University Hospital, San Sebastian, Spain
| | - María García-Martos
- Department of Pathology, Gregorio Marañon University Hospital, Madrid, Spain
| | - Ankur R Sangoi
- Department of Pathology, Stanford University, Stanford, CA, USA
| |
Collapse
|
11
|
Sun Y, Zhang X, Zhu H. Identify novel inflammation-related prognostic signature in pancreatic cancer patients. Medicine (Baltimore) 2024; 103:e36932. [PMID: 38363947 PMCID: PMC10869063 DOI: 10.1097/md.0000000000036932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 12/20/2023] [Indexed: 02/18/2024] Open
Abstract
Pancreatic cancer (PC) is a malignant tumor of the digestive system with a poor prognosis. PC patients with pancreatitis have a worse prognosis. But nobody reported the relationship between inflammation and prognosis in PC. Based on this, we are going to explore inflammation-related prognostic signature to predict patients' survival and potential therapeutic target. We screened gene expression profile and corresponding clinical information of patients from The Cancer Genome Atlas (TCGA) database. Gene set enrichment analysis (GSEA) was performed to identify differentially expressed genes (DEGs) between tumor and normal tissues with P value < .05. Univariate and multivariate Cox regression analysis was applied to identify possible prognostic inflammation genes and establish an inflammation-related risk score system, which was validated by Kaplan-Meier and Receiver operating characteristic (ROC) curves. Finally, we used the TISIDB database to predict targeted drugs for up-regulated gene hepatocyte growth factor receptor (MET) and used AUTODOCK software for molecular docking. We built a prognostic model consisted of 3 inflammation-related genes (tumor necrosis factor receptor associated factor 1/TFAR1, tyrosine kinase 2/TYK2, MET). According to the median value of those genes' risk score, PC patients were ranked into high- (88) and low-risk (89) groups. Then, the results of the Kaplan-Meier curves and the area under the curve (AUC) of the ROC curves showed this model had a good predictive power (P < .001, AUC = 0.806). The result of human protein atlas (HPA) database showed the expression of TRAF1 and TYK2 were low in pancreatic cancer, the expression of MET was high. TISIDB database founded brigatinib could target to MET. And AUTODOCK showed brigatinib had a nice docking with MET. Taken together, our study suggested that inflammation-associated prognostic signature might be used as novel biomarkers for predicting prognosis in PC patients and potential therapeutic target of the disease.
Collapse
Affiliation(s)
- Yuan Sun
- Department of Gastroenterology, the First Hospital of China Medical University, Shenyang, China
| | - Xiaoying Zhang
- Central Sterile Supply Department, the First Hospital of China Medical University, Shenyang, China
| | - Haiyan Zhu
- Department of Gastroenterology, the First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
12
|
Wang Y, Suarez ER, Kastrunes G, de Campos NSP, Abbas R, Pivetta RS, Murugan N, Chalbatani GM, D'Andrea V, Marasco WA. Evolution of cell therapy for renal cell carcinoma. Mol Cancer 2024; 23:8. [PMID: 38195534 PMCID: PMC10775455 DOI: 10.1186/s12943-023-01911-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 12/05/2023] [Indexed: 01/11/2024] Open
Abstract
Treatment for renal cell carcinoma (RCC) has improved dramatically over the last decade, shifting from high-dose cytokine therapy in combination with surgical resection of tumors to targeted therapy, immunotherapy, and combination therapies. However, curative treatment, particularly for advanced-stage disease, remains rare. Cell therapy as a "living drug" has achieved hematological malignancy cures with a high response rate, and significant research efforts have been made to facilitate its translation to solid tumors. Herein, we overview the cellular therapies for RCC focusing on allogeneic hematopoietic stem cell transplantation, T cell receptor gene-modified T cells, chimeric antigen receptor (CAR) T cells, CAR natural killer (NK) cells, lymphokine-activated killer (LAK) cells, γδ T cells, and dendritic cell vaccination. We have also included perspectives for using other recent approaches, such as CAR macrophages, dendritic cell-cytokine induced killer cells and regulatory CAR-T cells to shed light on preclinical development of cell therapy and advancing cell therapy into clinic to achieve cures for RCC.
Collapse
Affiliation(s)
- Yufei Wang
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
- Harvard Medical School, Boston, MA, 02215, USA
| | - Eloah Rabello Suarez
- Center for Natural and Human Sciences, Federal University of ABC, Santo Andre, SP, 09210-580, Brazil
- Graduate Program in Medicine - Hematology and Oncology, Federal University of Sao Paulo, São Paulo, SP, 04023-062, Brazil
| | - Gabriella Kastrunes
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Najla Santos Pacheco de Campos
- Center for Natural and Human Sciences, Federal University of ABC, Santo Andre, SP, 09210-580, Brazil
- Graduate Program in Medicine - Hematology and Oncology, Federal University of Sao Paulo, São Paulo, SP, 04023-062, Brazil
| | - Rabia Abbas
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Renata Schmieder Pivetta
- Center for Natural and Human Sciences, Federal University of ABC, Santo Andre, SP, 09210-580, Brazil
- Graduate Program in Medicine - Hematology and Oncology, Federal University of Sao Paulo, São Paulo, SP, 04023-062, Brazil
| | - Nithyassree Murugan
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | | | - Vincent D'Andrea
- Department of Surgery, Brigham and Women's Hospital, Boston, MA, 02215, USA
| | - Wayne A Marasco
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA.
- Harvard Medical School, Boston, MA, 02215, USA.
| |
Collapse
|
13
|
de Moraes FCA, Vilbert M, Alves VFC, de Oliveira Almeida G, Priantti JN, Madeira T, Stecca C, Fernandes MR, dos Santos NPC. Mesenchymal-Epithelial Transition Kinase Inhibitor Therapy in Patients with Advanced Papillary Renal-Cell Carcinoma: A Systematic Review and Meta-Analysis. Int J Mol Sci 2023; 24:17582. [PMID: 38139411 PMCID: PMC10744118 DOI: 10.3390/ijms242417582] [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: 10/07/2023] [Revised: 11/25/2023] [Accepted: 12/01/2023] [Indexed: 12/24/2023] Open
Abstract
Papillary subtypes of renal-cell carcinoma (pRCC) represent 10-15% of the cases and commonly have MET alterations. This systematic review and single-arm meta-analysis evaluated MET inhibitor therapy (METi) efficacy and safety in adults with confirmed advanced pRCC. The search strategy included PubMed, Web-of-science, Cochrane, and Scopus. We used the DerSimonian/Laird random effect model for all analyses; p-value < 5% was considered significant, and heterogeneity was assessed with I2. Three clinical trials and six cohort studies were included with 504 patients; 31% were MET-driven. Our pooled analysis demonstrated an objective response rate (ORR) in MET-driven, MET-independent, and overall patients of: 36% (95%CI: 10-62), 0% (95%CI: 0-3), and 21% (95%CI: 1-41), respectively. One-year disease control and progression-free survival rates were, respectively, 70% (95%CI: 52-88) and 15% (95%CI: 10-20). Twelve- and twenty-four-month survival rates were, respectively, 43% (95%CI: 23-64) and 10% (95%CI: 0-30). The prevalence of adverse events of any grade and grades 3-5 were 96% (95%CI: 91-100) and 44% (95%CI: 37-50), respectively. We suggest METi has anti-tumor activity and is tolerable in patients with advanced pRCC.
Collapse
Affiliation(s)
| | - Maysa Vilbert
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5T 2S8, Canada
| | | | | | - Jonathan N. Priantti
- School of Medicine, Federal University of Amazonas—UFAM, Manaus 69020-160, Brazil
| | - Thiago Madeira
- School of Medicine, Federal University of Minas Gerais—UFMG, Belo Horizonte 31270-901, Brazil
| | - Carlos Stecca
- Mackenzie Evangelical University Hospital, Curitiba 80710-390, Brazil
| | | | | |
Collapse
|
14
|
Chen YW, Wang L, Panian J, Dhanji S, Derweesh I, Rose B, Bagrodia A, McKay RR. Treatment Landscape of Renal Cell Carcinoma. Curr Treat Options Oncol 2023; 24:1889-1916. [PMID: 38153686 PMCID: PMC10781877 DOI: 10.1007/s11864-023-01161-5] [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] [Accepted: 11/28/2023] [Indexed: 12/29/2023]
Abstract
OPINION STATEMENT The treatment landscape of renal cell carcinoma (RCC) has evolved significantly over the past three decades. Active surveillance and tumor ablation are alternatives to extirpative therapy in appropriately selected patients. Stereotactic body radiation therapy (SBRT) is an emerging noninvasive alternative to treat primary RCC tumors. The advent of immune checkpoint inhibitors (ICIs) has greatly improved the overall survival of advanced RCC, and now the ICI-based doublet (dual ICI-ICI doublet; or ICI in combination with a vascular endothelial growth factor tyrosine kinase inhibitor, ICI-TKI doublet) has become the standard frontline therapy. Based on unprecedented outcomes in the metastatic with ICIs, they are also being explored in the neoadjuvant and adjuvant setting for patients with high-risk disease. Adjuvant pembrolizumab has proven efficacy to reduce the risk of RCC recurrence after nephrectomy. Historically considered a radioresistant tumor, SBRT occupies an expanding role to treat RCC with oligometastasis or oligoprogression in combination with systemic therapy. Furthermore, SBRT is being investigated in combination with ICI-doublet in the advanced disease setting. Lastly, given the treatment paradigm is shifting to adopt ICIs at earlier disease course, the prospective studies guiding treatment sequencing in the post-ICI setting is maturing. The effort is ongoing in search of predictive biomarkers to guide optimal treatment option in RCC.
Collapse
Affiliation(s)
- Yu-Wei Chen
- Division of Hematology Oncology, University of California San Diego, San Diego, CA, USA
| | - Luke Wang
- Department of Urology, University of California San Diego, San Diego, CA, USA
| | - Justine Panian
- School of Medicine, University of California San Diego, San Diego, CA, USA
| | - Sohail Dhanji
- Department of Urology, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Ithaar Derweesh
- Department of Urology, University of California San Diego, San Diego, CA, USA
| | - Brent Rose
- Department of Radiation Oncology, University of California San Diego, San Diego, CA, USA
| | - Aditya Bagrodia
- Department of Urology, University of California San Diego, San Diego, CA, USA
| | - Rana R McKay
- Division of Hematology Oncology, University of California San Diego, San Diego, CA, USA.
| |
Collapse
|
15
|
de Vries-Brilland M, Rioux-Leclercq N, Meylan M, Dauvé J, Passot C, Spirina-Menand E, Flippot R, Fromont G, Gravis G, Geoffrois L, Chevreau C, Rolland F, Blanc E, Lefort F, Ravaud A, Gross-Goupil M, Escudier B, Negrier S, Albiges L. Comprehensive analyses of immune tumor microenvironment in papillary renal cell carcinoma. J Immunother Cancer 2023; 11:e006885. [PMID: 37935564 PMCID: PMC10649801 DOI: 10.1136/jitc-2023-006885] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/19/2023] [Indexed: 11/09/2023] Open
Abstract
BACKGROUND Papillary renal cell carcinoma (pRCC) is the most common non-clear cell RCC, and associated with poor outcomes in the metastatic setting. In this study, we aimed to comprehensively evaluate the immune tumor microenvironment (TME), largely unknown, of patients with metastatic pRCC and identify potential therapeutic targets. METHODS We performed quantitative gene expression analysis of TME using Microenvironment Cell Populations-counter (MCP-counter) methodology, on two independent cohorts of localized pRCC (n=271 and n=98). We then characterized the TME, using immunohistochemistry (n=38) and RNA-sequencing (RNA-seq) (n=30) on metastatic pRCC from the prospective AXIPAP trial cohort. RESULTS Unsupervised clustering identified two "TME subtypes", in each of the cohorts: the "immune-enriched" and the "immune-low". Within AXIPAP trial cohort, the "immune-enriched" cluster was significantly associated with a worse prognosis according to the median overall survival to 8 months (95% CI, 6 to 29) versus 37 months (95% CI, 20 to NA, p=0.001). The two immune signatures, Teff and JAVELIN Renal 101 Immuno signature, predictive of response to immune checkpoint inhibitors (CPI) in clear cell RCC, were significantly higher in the "immune-enriched" group (adjusted p<0.05). Finally, five differentially overexpressed genes were identified, corresponding mainly to B lymphocyte populations. CONCLUSION For the first time, using RNA-seq and immunohistochemistry, we have highlighted a specific immune TME subtype of metastatic pRCC, significantly more infiltrated with T and B immune population. This "immune-enriched" group appears to have a worse prognosis and could have a potential predictive value for response to immunotherapy, justifying the confirmation of these results in a cohort of metastatic pRCC treated with CPI and in combination with targeted therapies. TRIAL REGISTRATION NUMBER NCT02489695.
Collapse
Affiliation(s)
- Manon de Vries-Brilland
- Department of Medical Oncology, Integrated Centers of Oncology (ICO) Paul Papin, Angers, France
| | | | - Maxime Meylan
- Equipe inflammation, complément et cancer, Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université Paris-Cité, Paris, France
| | - Jonathan Dauvé
- Department of Clinical Biology, Integrated Centers of Oncology (ICO) Paul Papin, Angers, France
| | - Christophe Passot
- Department of Clinical Biology, Integrated Centers of Oncology (ICO) Paul Papin, Angers, France
| | - Elena Spirina-Menand
- Department of Clinical Biology, Integrated Centers of Oncology (ICO) Paul Papin, Angers, France
| | - Ronan Flippot
- Medical Oncology, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | | | | | - Lionnel Geoffrois
- Department of Medical Oncology, Institut de Cancérologie de Lorraine, Vandoeuvre-les-Nancy, France
| | - Christine Chevreau
- Department of Medical Oncology, IUCT-Oncopôle Institut Claudius Regaud, Toulouse, France
| | - Fréderic Rolland
- Department of Medical Oncology, Integrated Centers of Oncology (ICO) René Gauducheau, Nantes, France
| | - Ellen Blanc
- Department of Clinical Research and Innovation, Centre Léon Bérard, Lyon, France
| | - Félix Lefort
- Department of Medical Oncology, University Hospital of Bordeaux, Bordeaux, France
| | - Alain Ravaud
- Department of Medical Oncology, University Hospital of Bordeaux, Bordeaux, France
| | - Marine Gross-Goupil
- Department of Medical Oncology, University Hospital of Bordeaux, Bordeaux, France
| | - Bernard Escudier
- Medical Oncology, Gustave Roussy, Université Paris-Saclay, Villejuif, France
- U1015 INSERM, Gustave Roussy Cancer Campus, Paris-Saclay University, Villejuif, France
| | - Sylvie Negrier
- Department of Medical Oncology, Lyon I University, Lyon, France
| | - Laurence Albiges
- Medical Oncology, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| |
Collapse
|
16
|
Semenescu LE, Kamel A, Ciubotaru V, Baez-Rodriguez SM, Furtos M, Costachi A, Dricu A, Tătăranu LG. An Overview of Systemic Targeted Therapy in Renal Cell Carcinoma, with a Focus on Metastatic Renal Cell Carcinoma and Brain Metastases. Curr Issues Mol Biol 2023; 45:7680-7704. [PMID: 37754269 PMCID: PMC10528141 DOI: 10.3390/cimb45090485] [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: 07/27/2023] [Revised: 09/07/2023] [Accepted: 09/19/2023] [Indexed: 09/28/2023] Open
Abstract
The most commonly diagnosed malignancy of the urinary system is represented by renal cell carcinoma. Various subvariants of RCC were described, with a clear-cell type prevailing in about 85% of all RCC tumors. Patients with metastases from renal cell carcinoma did not have many effective therapies until the end of the 1980s, as long as hormonal therapy and chemotherapy were the only options available. The outcomes were unsatisfactory due to the poor effectiveness of the available therapeutic options, but then interferon-alpha and interleukin-2 showed treatment effectiveness, providing benefits but only for less than half of the patients. However, it was not until 2004 that targeted therapies emerged, prolonging the survival rate. Currently, new technologies and strategies are being developed to improve the actual efficacy of available treatments and their prognostic aspects. This article summarizes the mechanisms of action, importance, benefits, adverse events of special interest, and efficacy of immunotherapy in metastatic renal cell carcinoma, with a focus on brain metastases.
Collapse
Affiliation(s)
- Liliana Eleonora Semenescu
- Department of Biochemistry, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, Str. Petru Rares nr. 2-4, 710204 Craiova, Romania; (L.E.S.); (A.C.)
| | - Amira Kamel
- Neurosurgical Department, Clinical Emergency Hospital “Bagdasar-Arseni”, Soseaua Berceni 12, 041915 Bucharest, Romania; (A.K.); (V.C.); (S.M.B.-R.); (L.G.T.)
| | - Vasile Ciubotaru
- Neurosurgical Department, Clinical Emergency Hospital “Bagdasar-Arseni”, Soseaua Berceni 12, 041915 Bucharest, Romania; (A.K.); (V.C.); (S.M.B.-R.); (L.G.T.)
| | - Silvia Mara Baez-Rodriguez
- Neurosurgical Department, Clinical Emergency Hospital “Bagdasar-Arseni”, Soseaua Berceni 12, 041915 Bucharest, Romania; (A.K.); (V.C.); (S.M.B.-R.); (L.G.T.)
| | - Mircea Furtos
- Neurosurgical Department, University Emergency Hospital of Bucharest, 050098 Bucharest, Romania;
| | - Alexandra Costachi
- Department of Biochemistry, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, Str. Petru Rares nr. 2-4, 710204 Craiova, Romania; (L.E.S.); (A.C.)
| | - Anica Dricu
- Department of Biochemistry, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, Str. Petru Rares nr. 2-4, 710204 Craiova, Romania; (L.E.S.); (A.C.)
| | - Ligia Gabriela Tătăranu
- Neurosurgical Department, Clinical Emergency Hospital “Bagdasar-Arseni”, Soseaua Berceni 12, 041915 Bucharest, Romania; (A.K.); (V.C.); (S.M.B.-R.); (L.G.T.)
- Department of Neurosurgery, Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania
| |
Collapse
|
17
|
Govindarajan A, Salgia NJ, Li H, Castro DV, Mirzapoiazova T, Armstrong B, Zhao D, Mercier BD, Dizman N, Chawla N, Zengin Z, Meza L, Tripathi N, Sayegh N, Chehrazi-Raffle A, Tripathi A, Pal SK. Characterization of papillary and clear cell renal cell carcinoma through imaging mass cytometry reveals distinct immunologic profiles. Front Immunol 2023; 14:1182581. [PMID: 37638025 PMCID: PMC10457014 DOI: 10.3389/fimmu.2023.1182581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 07/20/2023] [Indexed: 08/29/2023] Open
Abstract
Objective To characterize and further compare the immune cell populations of the tumor microenvironment (TME) in both clear cell and papillary renal cell carcinoma (RCC) using heavy metal-labeled antibodies in a multiplexed imaging approach (imaging mass cytometry). Materials and methods Formalin-fixed paraffin-embedded (FFPE) baseline tumor tissues from metastatic patients with clear cell renal cell carcinoma (ccRCC) and papillary renal cell carcinoma (pRCC) were retrospectively requisitioned from an institutional biorepository. Pretreated FFPE samples from 33 RCC patients (10 ccRCC, 23 pRCC) were accessioned and stained for imaging mass cytometry (IMC) analysis. Clinical characteristics were curated from an institutional RCC database. FFPE samples were prepared and stained with heavy metal-conjugated antibodies for IMC. An 11-marker panel of tumor stromal and immune markers was used to assess and quantify cellular relationships in TME compartments. To validate our time-of-flight (CyTOF) analysis, we cross-validated findings with The Cancer Genome Atlas Program (TCGA) analysis and utilized the CIBERSORTx tool to examine the abundance of main immune cell types in pRCC and ccRCC patients. Results Patients with ccRCC had a longer median overall survival than did those with pRCC (67.7 vs 26.8 mo, respectively). Significant differences were identified in the proportion of CD4+ T cells between disease subtypes (ccRCC 14.1%, pRCC 7.0%, p<0.01). Further, the pRCC cohort had significantly more PanCK+ tumor cells than did the ccRCC cohort (24.3% vs 9.5%, respectively, p<0.01). There were no significant differences in macrophage composition (CD68+) between cohorts. Our results demonstrated a significant correlation between the CyTOF and TCGA analyses, specifically validating that ccRCC patients exhibit higher levels of CD4+ T cells (ccRCC 17.60%, pRCC 15.7%, p<0.01) and CD8+ T cells (ccRCC 17.83%, pRCC 11.15%, p<0.01). The limitation of our CyTOF analysis was the large proportion of cells that were deemed non-characterizable. Conclusions Our findings emphasize the need to investigate the TME in distinct RCC histological subtypes. We observed a more immune infiltrative phenotype in the TME of the ccRCC cohort than in the pRCC cohort, where a tumor-rich phenotype was noted. As practical predictive biomarkers remain elusive across all subtypes of RCC, further studies are warranted to analyze the biomarker potential of such TME classifications.
Collapse
Affiliation(s)
- Ameish Govindarajan
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, CA, United States
| | - Nicholas J. Salgia
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, CA, United States
- Jacobs School of Medicine and Biomedical Sciences, Buffalo, NY, United States
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Haiqing Li
- Integrative Genome Core, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, United States
| | - Daniela V. Castro
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, CA, United States
| | - Tamara Mirzapoiazova
- Integrative Genome Core, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, United States
| | - Brian Armstrong
- Light Microscopy/Digital Imaging Core, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, United States
| | - Dan Zhao
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, CA, United States
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Benjamin D. Mercier
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, CA, United States
| | - Nazli Dizman
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, CA, United States
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, United States
| | - Neal Chawla
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, CA, United States
| | - Zeynep Zengin
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, CA, United States
| | - Luis Meza
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, CA, United States
| | - Nishita Tripathi
- Huntsman Cancer Institute-University of Utah Health Care, Salt Lake City, UT, United States
| | - Nicolas Sayegh
- Huntsman Cancer Institute-University of Utah Health Care, Salt Lake City, UT, United States
| | - Alex Chehrazi-Raffle
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, CA, United States
| | - Abhishek Tripathi
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, CA, United States
| | - Sumanta K. Pal
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, CA, United States
| |
Collapse
|
18
|
Heydt C, Ihle MA, Merkelbach-Bruse S. Overview of Molecular Detection Technologies for MET in Lung Cancer. Cancers (Basel) 2023; 15:cancers15112932. [PMID: 37296895 DOI: 10.3390/cancers15112932] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 05/22/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023] Open
Abstract
MET tyrosine kinase receptor pathway activation has become an important actionable target in solid tumors. Aberrations in the MET proto-oncogene, including MET overexpression, the activation of MET mutations, MET mutations that lead to MET exon 14 skipping, MET gene amplifications, and MET fusions, are known to be primary and secondary oncogenic drivers in cancer; these aberrations have evolved as predictive biomarkers in clinical diagnostics. Thus, the detection of all known MET aberrations in daily clinical care is essential. In this review, current molecular technologies for the detection of the different MET aberrations are highlighted, including the benefits and drawbacks. In the future, another focus will be on the standardization of detection technologies for the delivery of reliable, quick, and affordable tests in clinical molecular diagnostics.
Collapse
Affiliation(s)
- Carina Heydt
- Faculty of Medicine, Institute of Pathology, University Hospital Cologne, University of Cologne, Kerpener Str. 62, 50937 Cologne, Germany
| | - Michaela Angelika Ihle
- Faculty of Medicine, Institute of Pathology, University Hospital Cologne, University of Cologne, Kerpener Str. 62, 50937 Cologne, Germany
| | - Sabine Merkelbach-Bruse
- Faculty of Medicine, Institute of Pathology, University Hospital Cologne, University of Cologne, Kerpener Str. 62, 50937 Cologne, Germany
| |
Collapse
|
19
|
Ishihara H, Nemoto Y, Nakamura K, Tachibana H, Ikeda T, Fukuda H, Yoshida K, Kobayashi H, Iizuka J, Shimmura H, Hashimoto Y, Kondo T, Takagi T. Comparison of Outcomes Between Therapeutic Combinations Based on Immune Checkpoint Inhibitors or Tyrosine Kinase Inhibitor Monotherapy for First-Line Therapy of Patients with Advanced Renal Cell Carcinoma Outside of Clinical Trials: A Real-World Retrospective Multi-Institutional Study. Target Oncol 2023; 18:209-220. [PMID: 36941516 DOI: 10.1007/s11523-023-00956-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/18/2023] [Indexed: 03/23/2023]
Abstract
BACKGROUND Clinical trials have demonstrated the superior efficacy of immune checkpoint inhibitor (ICI)-based combination therapy over sunitinib, a multi-target tyrosine kinase inhibitor (TKI), in patients with advanced renal cell carcinoma. However, such benefits have not been elucidated in populations outside of clinical trials. METHODS We retrospectively evaluated data from 467 patients with advanced renal cell carcinoma who received ICI-based combination therapy or TKIs, as first-line therapy. Clinical outcome was compared between ICI-based combination therapy and TKIs in each population divided according to trial eligibility. RESULTS Among 152 patients treated with ICI-based combination therapy and 315 patients treated with TKIs, 76 (50.0%) and 156 (49.5%) were trial ineligible, respectively. Overall survival (p = 0.0072) and objective response rate (p < 0.0001) were significantly higher in ICI-based combination therapy than in TKIs, but progression-free survival was comparable (p = 0.681). In the trial-eligible population, overall survival was longer (p = 0.0906) and the objective response rate was significantly higher (p = 0.0124) in ICI-based combination therapy than in TKIs. In the trial-ineligible population, overall survival (p = 0.0208) and objective response rate (p = 0.0006) were significantly higher with ICI-based combination therapy than with TKIs. A multivariate analysis also showed that ICI-based combination therapy was independently associated with prolonged overall survival (hazard ratio, 0.47; p = 0.0016). Regardless of trial eligibility, progression-free survival did not differ between ICI-based combination therapy and TKIs (trial eligible: p = 0.287; trial ineligible: p = 0.0708). CONCLUSIONS The present study, using real-world data, provides evidence indicating the therapeutic benefit of ICI-based combination therapy over TKIs for advanced renal cell carcinoma was more statistically significant in the trial-ineligible population than in the trial-eligible population.
Collapse
Affiliation(s)
- Hiroki Ishihara
- Department of Urology, Tokyo Women's Medical University Adachi Medical Center, 4-33-1 Kouhoku, Adachi-ku, Tokyo, Japan.
| | - Yuki Nemoto
- Department of Urology, Tokyo Women's Medical University Adachi Medical Center, 4-33-1 Kouhoku, Adachi-ku, Tokyo, Japan
- Department of Urology, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, Japan
| | - Kazutaka Nakamura
- Department of Urology, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, Japan
- Department of Urology, Jyoban Hospital, Uenodai 57, Joban Kamiyunagayamachi, Iwaki, Fukushima, Japan
| | - Hidekazu Tachibana
- Department of Urology, Saiseikai Kazo Hospital, 1680 Kamitakayanagi, Kazo, Saitama, Japan
| | - Takashi Ikeda
- Department of Urology, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, Japan
| | - Hironori Fukuda
- Department of Urology, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, Japan
| | - Kazuhiko Yoshida
- Department of Urology, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, Japan
| | - Hirohito Kobayashi
- Department of Urology, Tokyo Women's Medical University Adachi Medical Center, 4-33-1 Kouhoku, Adachi-ku, Tokyo, Japan
| | - Junpei Iizuka
- Department of Urology, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, Japan
| | - Hiroaki Shimmura
- Department of Urology, Jyoban Hospital, Uenodai 57, Joban Kamiyunagayamachi, Iwaki, Fukushima, Japan
| | - Yasunobu Hashimoto
- Department of Urology, Saiseikai Kawaguchi General Hospital, 5-11-5 Nishikawaguchi, Kawaguchi, Saitama, Japan
| | - Tsunenori Kondo
- Department of Urology, Tokyo Women's Medical University Adachi Medical Center, 4-33-1 Kouhoku, Adachi-ku, Tokyo, Japan
| | - Toshio Takagi
- Department of Urology, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, Japan
| |
Collapse
|
20
|
Possible role of combined therapy targeting MET and pro-HGF activation for renal cell carcinoma: analysis by human HGF-producing SCID mice. Hum Cell 2023; 36:775-785. [PMID: 36708441 DOI: 10.1007/s13577-023-00857-y] [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: 11/24/2022] [Accepted: 01/10/2023] [Indexed: 01/29/2023]
Abstract
MET is a high-affinity receptor tyrosine kinase of HGF (hepatocyte growth factor). HGF is secreted as an inactive single-chain precursor (pro-HGF), which requires proteolytic activation for conversion to an active form. HGF activator inhibitor (HAI)-2 is a transmembrane Kunitz-type serine protease inhibitor, which inhibits all pro-HGF-activating enzymes. In RCC, increased expression of MET and decreased expression of HAI-2 were reported to be poor prognostic factors. In the current study, we tried to inhibit the growth of RCC cells by dual inhibition of both MET phosphorylation and pro-HGF-activation using MET inhibitor and HAI-2 overexpression. A transgenic mouse model which expressed human HGF (HGF mouse) was used for in vivo analysis to evaluate the HGF/MET signaling axis accurately. Initially, doxycycline-induced HAI-2 overexpression RCC cells (786-O-HAI2) were prepared. The cells were cultured with pro-HGF, and inhibitory effect of MET inhibitor (SCC244) and HAI-2 was evaluated by phosphorylation of MET and cell proliferation. Next, the cells were subcutaneously implanted to HGF mice and the growth inhibition was determined by SCC244 and HAI-2. Single use of each inhibitor showed significant inhibition in MET phosphorylation, migration and proliferation of 786-O-HAI2 cells; however, the strongest effect was observed by combined use of both inhibitors. Although in vivo analysis also showed apparent downregulation of MET phosphorylation and growth inhibition in combined treatment, statistical significance was not observed compared with single use of MET inhibitor. Combined treatment with MET-TKI and HAI-2 suggested to consider as a candidate for new strong therapy for RCC.
Collapse
|
21
|
Brown JT, Nazha B, Bilen MA. Combined Programmed Death-Ligand 1 and MET Inhibition: Has Papillary Renal Cell Carcinoma MET Its Match? J Clin Oncol 2023; 41:2467-2470. [PMID: 36809042 DOI: 10.1200/jco.22.02600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2023] Open
Affiliation(s)
- Jacqueline T Brown
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA.,Winship Cancer Institute of Emory University, Atlanta, GA
| | - Bassel Nazha
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA.,Winship Cancer Institute of Emory University, Atlanta, GA
| | - Mehmet Asim Bilen
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA.,Winship Cancer Institute of Emory University, Atlanta, GA
| |
Collapse
|
22
|
Chawla NS, Sayegh N, Prajapati S, Chan E, Pal SK, Chehrazi-Raffle A. An Update on the Treatment of Papillary Renal Cell Carcinoma. Cancers (Basel) 2023; 15:565. [PMID: 36765524 PMCID: PMC9913225 DOI: 10.3390/cancers15030565] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 01/11/2023] [Accepted: 01/15/2023] [Indexed: 01/19/2023] Open
Abstract
Papillary renal cell carcinoma (pRCC) is the second-most common subtype of kidney cancer following clear cell renal cell carcinoma (ccRCC), representing 15% of kidney cancers. Despite advances in therapy, including combination strategies with targeted therapies and immune checkpoint inhibitors, progress has lagged behind that of ccRCC. This is in part due to the heterogenous nature of the various subtypes of pRCC. More recently, investigators have turned efforts towards histology and biology-based trials. In this review, we outline some of the distinct biological characteristics of pRCC and discuss the most impactful clinical trials to date. Finally, we look ahead to several highly anticipated ongoing trials in pRCC.
Collapse
Affiliation(s)
- Neal S. Chawla
- Department of Medical Oncology & Experimental Therapeutics, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Nicolas Sayegh
- Division of Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Sweta Prajapati
- Department of Medical Oncology & Experimental Therapeutics, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Elyse Chan
- Department of Medical Oncology & Experimental Therapeutics, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Sumanta K. Pal
- Department of Medical Oncology & Experimental Therapeutics, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Alexander Chehrazi-Raffle
- Department of Medical Oncology & Experimental Therapeutics, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| |
Collapse
|
23
|
Xie T, Liu B, Liu D, Zhou Y, Yang Q, Wang D, Tang M, Liu W. Cuproptosis-related lncRNA signatures predict prognosis and immune relevance of kidney renal papillary cell carcinoma. Front Pharmacol 2022; 13:1103986. [PMID: 36618928 PMCID: PMC9810632 DOI: 10.3389/fphar.2022.1103986] [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: 11/21/2022] [Accepted: 12/12/2022] [Indexed: 12/24/2022] Open
Abstract
Kidney renal papillary cell carcinoma (KIRP) has a high mortality rate and a poor prognosis. Cu concentrations differed significantly between renal cancer tissues and adjacent normal tissues. Cuproptosis is a newly identified cell death. Long non-coding RNAs (lncRNAs) play a crucial role in the progression of KIRP. In this study, we focused on constructing and validating cuproptosis-related lncRNA signatures to predict the prognosis of KIRP patients and their immune correlation. We created prognosis models using Cox regression analysis and the least absolute shrinkage and selection operator (LASSO) algorithm. We found that patients in the high-risk group had poorer overall survival (OS) and progression-free survival (PFS) and higher mortality. Risk score and stage are prognosis factors independent of other clinical features. Kaplan-Meier analysis, receiver operating characteristic (ROC) curves, and C-index curves showed that cuproptosis-related lncRNA signatures could more accurately predict the prognosis of patients. Functional enrichment analysis suggests that the function of differentially expressed genes (DEGs) is associated with KIRP development and immunity. In immune-related function analysis, we found a significant difference in parainflammation responses between high-risk and low-risk groups. The mutation frequencies of TTN, MET, KMT2C, PKHD1, SETD2, and KMT2D genes in the high-risk group were higher than those in the low-risk group, but the mutation frequencies of MUC16, KIAA109, CUBN, USH2A, DNAH8 and HERC2 genes were significantly lower than those in the low-risk group. Survival analysis of tumor mutation burden (TMB) and combined TMB-risk showed better OS in patients with high TMB. Immune infiltration and immune checkpoint analysis assessed the immune association of six high mutation frequency genes (TTN, MET, KMT2C, PKHD1, SETD2, and KMT2D) with KIRP. Finally, we performed a drug sensitivity analysis and screened 15 potential drugs that differed between high-risk and low-risk patients. In this study, we constructed and validated cuproptosis-related lncRNA signatures that can more accurately predict the prognosis of KIRP patients and provide new potential therapeutic targets and prognosis markers for KIRP patients.
Collapse
Affiliation(s)
- Tongjin Xie
- Department of Urology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Bin Liu
- Department of Urology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Dongbo Liu
- Department of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Yusong Zhou
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Qingping Yang
- Department of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Dai Wang
- Xiangya School of Pharmacy, Central South University, Changsha, China
| | - Mengjie Tang
- Department of Pathology, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Wei Liu
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, China,*Correspondence: Wei Liu,
| |
Collapse
|
24
|
Thouvenin J, Alhalabi O, Carlo M, Carril-Ajuria L, Hirsch L, Martinez-Chanza N, Négrier S, Campedel L, Martini D, Borchiellini D, Chahoud J, Lodi M, Barthélémy P, Hasanov E, Hahn AW, Gil T, Viswanathan SR, Bakouny Z, Msaouel P, Asim Bilen M, Choueiri TK, Albiges L, Tannir NM, Malouf GG. Efficacy of Cabozantinib in Metastatic MiT Family Translocation Renal Cell Carcinomas. Oncologist 2022; 27:1041-1047. [PMID: 35979929 DOI: 10.1093/oncolo/oyac158] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Accepted: 06/24/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND MiT family translocation renal cell carcinoma (TRCC) is a rare and aggressive subgroup of renal cell carcinoma harboring high expression of c-MET. While TRCC response rates to VEGF receptor tyrosine kinase inhibitors (TKIs) and immune checkpoint inhibitors are limited, efficacy of cabozantinib (a VEGFR, MET, and AXL inhibitor) in this subgroup is unclear. METHODS We performed a multicenter, retrospective, international cohort study of patients with TRCC treated with cabozantinib. The main objectives were to estimate response rate according to RECIST 1.1 and to analyze progression-free survival (PFS) and overall survival (OS). RESULTS Fifty-two patients with metastatic TRCC treated in the participating centers and evaluable for response were included. Median age at metastatic diagnosis was 40 years (IQR 28.5-53). Patients' IMDC risk groups at diagnosis were favorable (9/52), intermediate (35/52), and poor (8/52). Eleven (21.2%) patients received cabozantinib as frontline therapy, 15 (28.8%) at second line, and 26 (50%) at third line and beyond. The proportion of patients who achieved an objective response was 17.3%, including 2 complete responses and 7 partial responses. For 26 (50%) patients, stable disease was the best response. With a median follow-up of 25.1 months (IQR 12.6-39), median PFS was 6.8 months (95%CI 4.6-16.3) and median OS was 18.3 months (95%CI 17.0-30.6). No difference of response was identified according to fusion transcript features. CONCLUSION This real-world study provides evidence of the activity of cabozantinib in TRCC, with more durable responses than those observed historically with other VEGFR-TKIs or ICIs.
Collapse
Affiliation(s)
- Jonathan Thouvenin
- Institut de Cancérologie Strasbourg Europe (ICANS/HUS), Strasbourg, France.,Institut de Cancérologie des Hospices Civils de Lyon, Lyon, France.,Dana-Farber Cancer Institute (DFCI), Boston, MA, USA
| | | | - Maria Carlo
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Laure Hirsch
- Dana-Farber Cancer Institute (DFCI), Boston, MA, USA
| | | | - Sylvie Négrier
- Université Claude Bernard, Centre Léon Bérard, Lyon, France
| | - Luca Campedel
- AP-HP, Groupe hospitalier Pitié-Salpêtrière, Paris, France
| | - Dylan Martini
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | | | - Jad Chahoud
- Department of Genitourinary Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Massimo Lodi
- Institut de Cancérologie Strasbourg Europe (ICANS/HUS), Strasbourg, France
| | | | | | | | | | | | - Ziad Bakouny
- Dana-Farber Cancer Institute (DFCI), Boston, MA, USA
| | | | | | | | | | | | - Gabriel G Malouf
- Institut de Cancérologie Strasbourg Europe (ICANS/HUS), Strasbourg, France
| |
Collapse
|
25
|
Benjamin DJ, Nolla K, Cella D. Nivolumab plus cabozantinib for advanced renal cell carcinoma. Future Oncol 2022; 18:4351-4359. [PMID: 36655774 DOI: 10.2217/fon-2022-0785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Advanced renal cell carcinoma (RCC) remains a challenging oncologic disease to treat despite advancements in therapeutics. Nonetheless, the development of tyrosine kinase inhibitors (TKIs) and immunotherapy has drastically altered the treatment landscape for advanced RCC over the past decade. The current standard-of-care treatment for advanced RCC involves combination TKI and immunotherapy regimens including cabozantinib and nivolumab as studied in the CheckMate 9ER trial. This review summarizes the preclinical and clinical evidence that led to the CheckMate 9ER study, as well as pertinent study aspects such as treatment efficacy, adverse events and patient-related outcomes.
Collapse
Affiliation(s)
- David J Benjamin
- Hoag Family Cancer Institute, 1 Hoag Drive, Building 51, Newport Beach, CA 92663, USA
| | - Kyle Nolla
- Department of Medical Social Sciences, Northwestern University Feinberg School of Medicine, 625 N. Michigan Avenue, Chicago, IL 60611, USA
| | - David Cella
- Department of Medical Social Sciences, Northwestern University Feinberg School of Medicine, 625 N. Michigan Avenue, Chicago, IL 60611, USA
| |
Collapse
|
26
|
Tyler LC, Le AT, Chen N, Nijmeh H, Bao L, Wilson TR, Chen D, Simmons B, Turner KM, Perusse D, Kasibhatla S, Christiansen J, Dudek AZ, Doebele RC. MET gene amplification is a mechanism of resistance to entrectinib in ROS1+ NSCLC. Thorac Cancer 2022; 13:3032-3041. [PMID: 36101520 PMCID: PMC9626307 DOI: 10.1111/1759-7714.14656] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 08/29/2022] [Accepted: 08/30/2022] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND ROS1 tyrosine kinase inhibitors (TKIs) have demonstrated significant clinical benefit for ROS1+ NSCLC patients. However, TKI resistance inevitably develops through ROS1 kinase domain (KD) modification or another kinase driving bypass signaling. While multiple TKIs have been designed to target ROS1 KD mutations, less is known about bypass signaling in TKI-resistant ROS1+ lung cancers. METHODS Utilizing a primary, patient-derived TPM3-ROS1 cell line (CUTO28), we derived an entrectinib-resistant line (CUTO28-ER). We evaluated proliferation and signaling responses to TKIs, and utilized RNA sequencing, whole exome sequencing, and fluorescence in situ hybridization to detect transcriptional, mutational, and copy number alterations, respectively. We substantiated in vitro findings using a CD74-ROS1 NSCLC patient's tumor samples. Last, we analyzed circulating tumor DNA (ctDNA) from ROS1+ NSCLC patients in the STARTRK-2 entrectinib trial to determine the prevalence of MET amplification. RESULTS CUTO28-ER cells did not exhibit ROS1 KD mutations. MET TKIs inhibited proliferation and downstream signaling and MET transcription was elevated in CUTO28-ER cells. CUTO28-ER cells displayed extrachromosomal (ecDNA) MET amplification without MET activating mutations, exon 14 skipping, or fusions. The CD74-ROS1 patient samples illustrated MET amplification while receiving ROS1 TKI. Finally, two of 105 (1.9%) entrectinib-resistant ROS1+ NSCLC STARTRK-2 patients with ctDNA analysis at enrollment and disease progression displayed MET amplification. CONCLUSIONS Treatment with ROS1-selective inhibitors may lead to MET-mediated resistance. The discovery of ecDNA MET amplification is noteworthy, as ecDNA is associated with more aggressive cancers. Following progression on ROS1-selective inhibitors, MET gene testing and treatments targeting MET should be explored to overcome MET-driven resistance.
Collapse
Affiliation(s)
- Logan C. Tyler
- Department of Medicine—Division of Medical OncologyUniversity of Colorado—Anschutz Medical CampusAuroraColoradoUSA
| | - Anh T. Le
- Department of Medicine—Division of Medical OncologyUniversity of Colorado—Anschutz Medical CampusAuroraColoradoUSA
| | - Nan Chen
- Department of Medicine—Division of Medical OncologyUniversity of Colorado—Anschutz Medical CampusAuroraColoradoUSA
| | - Hala Nijmeh
- Department of PathologyUniversity of Colorado—Anschutz Medical CampusAuroraColoradoUSA
| | - Liming Bao
- Department of PathologyUniversity of Colorado—Anschutz Medical CampusAuroraColoradoUSA
| | | | - David Chen
- Genentech, Inc.South San FranciscoCaliforniaUSA
| | | | | | | | | | | | - Arkadiusz Z. Dudek
- HealthPartners Cancer Center at Regions HospitalSt. PaulMinnesotaUSA,Department of Medicine—Division of Hematology, Oncologyand Transplantation University of MinnesotaMinneapolisMinnesotaUSA
| | - Robert C. Doebele
- Department of Medicine—Division of Medical OncologyUniversity of Colorado—Anschutz Medical CampusAuroraColoradoUSA
| |
Collapse
|
27
|
Biphasic squamoid alveolar papillary renal cell carcinoma: A unique papillary renal cell carcinoma with distinctive morphology, immunophenotype and molecular genetic features. Pathol Res Pract 2022; 239:154120. [DOI: 10.1016/j.prp.2022.154120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 07/28/2022] [Accepted: 09/08/2022] [Indexed: 11/20/2022]
|
28
|
Zschäbitz S, Mikuteit M, Stöhr C, Herrmann E, Polifka I, Agaimy A, Trojan L, Ströbel P, Becker F, Wülfing C, Barth P, Stöckle M, Staehler M, Stief C, Haferkamp A, Hohenfellner M, Duensing S, Macher-Göppinger S, Wullich B, Noldus J, Brenner W, Roos FC, Walter B, Otto W, Burger M, Schrader AJ, Hartmann A, Erlmeier F, Steffens S. Expression of nectin-4 in papillary renal cell carcinoma. Discov Oncol 2022; 13:90. [PMID: 36136143 PMCID: PMC9500133 DOI: 10.1007/s12672-022-00558-2] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 09/13/2022] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND Nectin-4 contributes to tumor proliferation, lymphangiogenesis and angiogenesis in malignant tumors and is an emerging target in tumor therapy. In renal cell carcinoma (RCC) VEGF-directed tyrosine kinase inhibitors and checkpoint inhibitors are currently treatments of choice. Enfortumab vedotin-ejf (EV) is an antibody drug conjugate that targets Nectin-4. The aim of our study was to investigate the expression of Nectin-4 in a large cohort of papillary RCC specimens. PATIENTS AND METHODS Specimens were derived from the PANZAR consortium (Erlangen, Heidelberg, Herne, Homburg, Mainz, Mannheim, Marburg, Muenster, LMU Munich, TU Munich, and Regensburg). Clinical data and tissue samples from n = 190 and n = 107 patients with type 1 and 2 pRCC, respectively, were available. Expression of Nectin-4 was determined by immunohistochemistry (IHC). RESULTS In total, Nectin-4 staining was moderately or strongly positive in of 92 (48.4%) of type 1 and 39 (36.4%) type 2 of pRCC cases. No associations between Nectin-4 expression and age at diagnosis, gender, grading, and TNM stage was found. 5 year overall survival rate was not statistically different in patients with Nectin-4 negative versus Nectin-4 positive tumors for the overall cohort and the pRCC type 2 subgroup, but higher in patient with Nectin-4 positive pRCC type 1 tumors compared to Nectin-4 negative tumors (81.3% vs. 67.8%, p = 0.042). CONCLUSION Nectin-4 could not be confirmed as a prognostic marker in pRCC in general. Due to its high abundance on pRCC specimens Nectin-4 is an interesting target for therapeutical approaches e.g. with EV. Clinical trials are warranted to elucidate its role in the pRCC treatment landscape.
Collapse
Affiliation(s)
- Stefanie Zschäbitz
- Dept. of Medical Oncology, National Center of Tumor Diseases, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Marie Mikuteit
- Department of Rheumatology and Immunology, Hanover Medical School, Carl-Neuberg-Straße 1, 30625 Hannover, Germany
- Dean’s Office – Curriculum Development, Hanover Medical School, 30625 Hannover, Germany
| | - Christine Stöhr
- Institute of Pathology, University Hospital Erlangen-Nuernberg, Friedrich Alexander University (FAU), 91054 Erlangen, Germany
| | - Edwin Herrmann
- Department of Urology, University Hospital Muenster, 48149 Muenster, Germany
- Present Address: Institute of Urology, Prosper-Hospital GmbH, 45659 Recklinghausen, Germany
| | - Iris Polifka
- Institute of Pathology, University Hospital Erlangen-Nuernberg, Friedrich Alexander University (FAU), 91054 Erlangen, Germany
| | - Abbas Agaimy
- Institute of Pathology, University Hospital Erlangen-Nuernberg, Friedrich Alexander University (FAU), 91054 Erlangen, Germany
| | - Lutz Trojan
- Department of Urology, University Hospital Göttingen, 37075 Göttingen, Germany
| | - Philipp Ströbel
- Department of Pathology, University Hospital Göttingen, 37075 Göttingen, Germany
| | - Frank Becker
- Department of Urology and Pediatric Urology, University of Saarland (UKS), 66421 Homburg, Germany
- Present Address: Urological Group and Clinic Derouet/Pönicke/Becker, Boxberg Centre, 66538 Neunkirchen, Germany
| | - Christian Wülfing
- Department of Urology, University Hospital Muenster, 48149 Muenster, Germany
- Present Address: Department of Urology, Asklepios Clinics Altona, 22763 Hamburg, Germany
| | - Peter Barth
- Department of Urology, University of Marburg, 35037 Marburg, Germany
- Present Address: Institute of Pathology/Gerhard-Domagk Institute, University Hospital Muenster, 48149 Muenster, Germany
| | - Michael Stöckle
- Department of Urology and Pediatric Urology, University of Saarland (UKS), 66421 Homburg, Germany
| | - Michael Staehler
- Department of Urology, University Hospital Munich, 81337 Munich, Germany
| | - Christian Stief
- Department of Urology, University Hospital Munich, 81337 Munich, Germany
| | - Axel Haferkamp
- Department of Urology, University Hospital Mainz, 55131 Mainz, Germany
- Present Address: Department of Urology and Pediatric Urology, University Hospital Mainz, 55131 Mainz, Germany
| | - Markus Hohenfellner
- Department of Urology, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Stefan Duensing
- Department of Urology, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | | | - Bernd Wullich
- Department of Urology and Pediatric Urology, University Hospital Erlangen-Nuernberg, Friedrich Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Joachim Noldus
- Department of Urology, Marien Hospital Herne, Ruhr University Bochum, 44625 Herne, Germany
| | - Walburgis Brenner
- Department of Urology, University Hospital Mainz, 55131 Mainz, Germany
- Present Address: Department of Gynecology, University of Mainz, 55131 Mainz, Germany
| | - Frederik C. Roos
- Department of Urology, University Hospital Frankfurt, 60590 Frankfurt, Germany
| | - Bernhard Walter
- Department of Urology and Pediatric Urology, University Hospital Erlangen-Nuernberg, Friedrich Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany
- Present Address: Department of Urology, Kreiskliniken Altötting-Burghausen, 84489 Burghausen, Germany
| | - Wolfgang Otto
- Department of Urology, Caritas St. Josef and University, 93053 Regensburg, Germany
| | - Maximilian Burger
- Department of Urology, Caritas St. Josef and University, 93053 Regensburg, Germany
| | - Andres Jan Schrader
- Department of Urology, University Hospital Muenster, 48149 Muenster, Germany
- Present Address: Department of Rheumatology and Immunology, Medical School Hannover, 30625 Hannover, Germany
| | - Arndt Hartmann
- Institute of Pathology, University Hospital Erlangen-Nuernberg, Friedrich Alexander University (FAU), 91054 Erlangen, Germany
| | - Franziska Erlmeier
- Institute of Pathology, University Hospital Erlangen-Nuernberg, Friedrich Alexander University (FAU), 91054 Erlangen, Germany
| | - Sandra Steffens
- Department of Rheumatology and Immunology, Hanover Medical School, Carl-Neuberg-Straße 1, 30625 Hannover, Germany
- Dean’s Office – Curriculum Development, Hanover Medical School, 30625 Hannover, Germany
- Present Address: Department of Rheumatology and Immunology, Hanover Medical School, 30625 Hannover, Germany
| |
Collapse
|
29
|
Albiges L, Heng DYC, Lee JL, Walker S, Mellemgaard A, Ottesen L, Frigault MM, L'Hernault A, Wessen J, Choueiri T, Cancel M, Signoretti S. Impact of MET status on treatment outcomes in papillary renal cell carcinoma: A pooled analysis of historical data. Eur J Cancer 2022; 170:158-168. [PMID: 35640484 DOI: 10.1016/j.ejca.2022.04.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 04/14/2022] [Indexed: 11/03/2022]
Abstract
BACKGROUND Papillary renal cell carcinoma (PRCC) represents 15% of RCCs but has no indicated therapies, with limited biomarker-based data to inform targeted treatment. MET alterations may be key; > 80% of PRCC tumours show MET upregulation. The objective of this study was to assess MET status in PRCC and its impact on clinical outcomes. METHODS This retrospective, observational study included patients with locally advanced/metastatic PRCC from three international registries. MET status was determined retrospectively by next generation sequencing (NGS) of archival tissue. MET-driven was defined as MET and/or hepatocyte growth factor amplification, chromosome 7 gain, and/or MET kinase domain mutations. Objectives included progression-free survival (PFS) and overall survival (OS) by MET status using a Cox proportional hazards model. RESULTS Of 308 patients, 305 received first-line treatment; most commonly sunitinib (n = 208; 68%), then everolimus (n = 40; 13%). Of 179 patients with valid NGS results, 38% had MET-driven and 49% MET-independent tumours (13% unevaluable). In the MET-driven versus MET-independent subgroups, respectively, of sunitinib-treated patients, median PFS was numerically longer, though not statistically significantly; PFS: 9.2 months (95% confidence interval [CI]: 5.4-13.2) versus 5.7 months (95% CI: 4.3-7.4), hazard ratio (HR) = 0.67; 95% CI: 0.41-1.08. There was no difference between the OS of each subgroup. CONCLUSIONS MET-driven PRCC may respond to targeted agents. However, the presence of MET alterations did not appear to be predictive for outcomes in response to current therapies, which are not biomarker-driven, compared with MET-independent tumours.
Collapse
Affiliation(s)
- Laurence Albiges
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Cancer Medicine, Institut Gustave Roussy, Université Paris Saclay, Villejuif, France.
| | - Daniel Y C Heng
- Department of Medical Oncology, Tom Baker Cancer Center, University of Calgary, Calgary, Alberta, Canada.
| | - Jae Lyun Lee
- Department of Oncology, Asan Medical Center and University of Ulsan College of Medicine, Seoul, South Korea.
| | | | | | | | | | - Anne L'Hernault
- Precision Medicine and Biosamples, Oncology R&D, AstraZeneca, Cambridge, UK.
| | | | - Toni Choueiri
- Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA.
| | - Mathilde Cancel
- Department of Medical Oncology, CHU Bretonneau Centre, University of Tours, Tours, France.
| | - Sabina Signoretti
- Harvard Medical School, Boston, MA, USA; Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA.
| |
Collapse
|
30
|
Abstract
PURPOSE OF THE REVIEW Papillary renal cell carcinoma (pRCC) is the second most frequent renal cancer subtype and represents 15-20% of all RCC. Classification of pRCC is changing because novel tumour entities have been discovered in the last years. In this review, we summarise recent studies relevant for the understanding of the molecular complexity and the broader differential diagnosis of pRCC. RECENT FINDINGS It has been 25 years ago, that pRCC was morphologically subdivided into type 1 and type 2. Recently described tumour entities in the 2022 WHO classification challenged this concept and allow a new view on the molecular background in pRCC. Biphasic hyalinizing psammomatous RCC and papillary renal neoplasm with reversed polarity are emerging tumour entities derived from the new concept of molecularly defined RCC subtypes. Immune checkpoint inhibition and tyrosine kinase inhibitors have been introduced as the new backbone in the first-line treatment of advanced pRCCs. To identify novel targeted treatments for patients with pRCC it is crucial to investigate the specific molecular background of pRCC considering emerging pRCC subtypes. SUMMARY In the future, a deeper understanding of the correlation between molecular aberrations and new pRCC subtypes may improve the classification of pRCC patients and could reveal potential predictive biomarkers for each subgroup.
Collapse
Affiliation(s)
- Silvia Angori
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
| | - João Lobo
- Department of Pathology, Portuguese Oncology Institute of Porto (IPOP)
- Cancer Biology and Epigenetics Group, IPO Porto Research Center (GEBC CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto) & Porto Comprehensive Cancer Center (P.CCC), R. Dr António Bernardino de Almeida
- Department of Pathology and Molecular Immunology, ICBAS–School of Medicine and Biomedical Sciences, University of Porto (ICBAS-UP), Porto, Portugal
| | - Holger Moch
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
- Faculty of Medicine, University of Zurich, Zurich, Switzerland
| |
Collapse
|
31
|
Srivastava A, Doppalapudi SK, Patel HV, Srinivasan R, Singer EA. The roaring 2020s: a new decade of systemic therapy for renal cell carcinoma. Curr Opin Oncol 2022; 34:234-242. [PMID: 35266906 PMCID: PMC9177746 DOI: 10.1097/cco.0000000000000831] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW The genomic and immunologic profiling of renal cell carcinoma (RCC) has provided the impetus for advancements in systemic treatments using combination therapy - either with immune check point inhibitor (ICI) + ICI or with ICI + targeted therapy. This approach has been examined in several landmark trials, treating both clear cell (ccRCC) and nonclear cell (nccRCC) histologies. In this review, we highlight systemic therapy advancements made in this new decade, the 2020s. RECENT FINDINGS Targeting the programmed death receptor 1/PD-L1 pathway has created more tolerable and effective immunotherapy regimens, expanding the applications of ICIs. These new applications, paired with trial data, include ICI monotherapy in nccRCC and adjuvant pembrolizumab in resected, high-risk RCC. In addition, ICI + ICI and ICI + TKI combination therapy have demonstrated oncologic efficacy in advanced ccRCC and sarcomatoid RCC. Similar progress has been noted regarding new targeted therapies. Along the hypoxia inducible factor pathway, belzutifan has received FDA approval in von Hippel-Lindau-associated RCC. In addition, in papillary RCC, agents such as cabozantinib target the MET proto-oncogene pathway and have demonstrated impressive oncologic outcomes. SUMMARY The 2020s utilize the molecular profiling of advanced RCC as a scaffold for recent trials in immunotherapy and targeted therapies. Going forward, emphasizing patient-reported outcomes and careful clinical trial construction remain critical to improve systemic therapy in RCC.
Collapse
Affiliation(s)
- Arnav Srivastava
- Section of Urologic Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey
| | | | - Hiren V. Patel
- Section of Urologic Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey
| | - Ramaprasad Srinivasan
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Eric A. Singer
- Section of Urologic Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey
| |
Collapse
|
32
|
Beck P, Selle B, Madenach L, Jones DTW, Vokuhl C, Gopisetty A, Nabbi A, Brecht IB, Ebinger M, Wegert J, Graf N, Gessler M, Pfister SM, Jäger N. The genomic landscape of pediatric renal cell carcinomas. iScience 2022; 25:104167. [PMID: 35445187 PMCID: PMC9014386 DOI: 10.1016/j.isci.2022.104167] [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: 11/16/2021] [Revised: 03/03/2022] [Accepted: 03/24/2022] [Indexed: 12/08/2022] Open
Abstract
Pediatric renal cell carcinomas (RCC) differ from their adult counterparts not only in histologic subtypes but also in clinical characteristics and outcome. However, the underlying biology is still largely unclear. For this reason, we performed whole-exome and transcriptome sequencing analyses on a cohort of 25 pediatric RCC patients with various histologic subtypes, including 10 MiT family translocation (MiT) and 10 papillary RCCs. In this cohort of pediatric RCC, we find only limited genomic overlap with adult RCC, even within the same histologic subtype. Recurrent somatic mutations in genes not previously reported in RCC were detected, such as in CCDC168, PLEKHA1, VWF, and MAP3K9. Our papillary pediatric RCCs, which represent the largest cohort to date with comprehensive molecular profiling in this age group, appeared as a distinct genomic subtype differing in terms of gene mutations and gene expression patterns not only from MiT-RCC but also from their adult counterparts. WES and RNA-seq of 25 pediatric RCCs with various histologic subtypes Detected only limited genomic overlap with adult RCC Revealed recurrent somatic mutations in genes not previously reported in RCC Discovery of a CRK-PITPNA fusion gene in a pediatric papillary RCC
Collapse
Affiliation(s)
- Pengbo Beck
- Hopp Children's Cancer Center Heidelberg (KiTZ) & Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany.,Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Barbara Selle
- Hopp Children's Cancer Center Heidelberg (KiTZ) & Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Lukas Madenach
- Hopp Children's Cancer Center Heidelberg (KiTZ) & Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany.,Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - David T W Jones
- Hopp Children's Cancer Center Heidelberg (KiTZ) & Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany.,Pediatric Glioma Research Group, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Christian Vokuhl
- Section of Pediatric Pathology, Department of Pathology, University Hospital Bonn, Bonn, Germany
| | - Apurva Gopisetty
- Hopp Children's Cancer Center Heidelberg (KiTZ) & Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany.,Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Arash Nabbi
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Ines B Brecht
- Department of Pediatric Oncology and Hematology, University Children's Hospital Tübingen, Tübingen, Germany
| | - Martin Ebinger
- Department of Pediatric Oncology and Hematology, University Children's Hospital Tübingen, Tübingen, Germany
| | - Jenny Wegert
- Theodor-Boveri-Institute/Biocenter, Developmental Biochemistry, Würzburg University & Comprehensive Cancer Center Mainfranken, Würzburg, Germany
| | - Norbert Graf
- Department of Pediatric Oncology and Hematology, Saarland University, Homburg, Germany
| | - Manfred Gessler
- Theodor-Boveri-Institute/Biocenter, Developmental Biochemistry, Würzburg University & Comprehensive Cancer Center Mainfranken, Würzburg, Germany
| | - Stefan M Pfister
- Hopp Children's Cancer Center Heidelberg (KiTZ) & Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany.,Department of Pediatric Oncology, Hematology and Immunology, University Hospital Heidelberg, Heidelberg, Germany
| | - Natalie Jäger
- Hopp Children's Cancer Center Heidelberg (KiTZ) & Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
| |
Collapse
|
33
|
Activity of ALK Inhibitors in Renal Cancer with ALK Alterations: A Systematic Review. Int J Mol Sci 2022; 23:ijms23073995. [PMID: 35409355 PMCID: PMC8999731 DOI: 10.3390/ijms23073995] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 03/30/2022] [Accepted: 03/31/2022] [Indexed: 02/06/2023] Open
Abstract
Renal cell carcinoma (RCC) associated with anaplastic lymphoma kinase (ALK) gene rearrangements (ALK-RCC) is currently considered an “emerging or provisional” tumor entity by the last World Health Organization classification published in 2016. Although several studies assessing ALK-RCC’s clinical and histological characteristics have been published in recent years, only a few publications have evaluated the activity of ALK inhibitors (ALK-i) in this subgroup of patients. Considering the well-recognized efficacy of this evolving class of targeted therapies in other ALK-positive tumors, we conducted a systematic review to evaluate the reported activity of ALK-i in the ALK-RCC subtype. MEDLINE was searched from its inception to 7 January 2022 for case reports and case series on adult metastatic ALK-RCC patients treated with ALK-i whose therapeutic outcomes were available. A virtual cohort of ALK-RCC patients was created. Our results showed a favorable activity of first- and second-generation ALK-i in pretreated ALK-RCC patients in terms of either radiological response or performance status improvement. We hope that the present work will prompt the creation of large, multi-institutional clinical trials to confirm these promising early data.
Collapse
|
34
|
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
|
35
|
Yang X, Liao HY, Zhang HH. Roles of MET in human cancer. Clin Chim Acta 2021; 525:69-83. [PMID: 34951962 DOI: 10.1016/j.cca.2021.12.017] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 12/15/2021] [Accepted: 12/17/2021] [Indexed: 01/18/2023]
Abstract
The MET proto-oncogene was first identified in osteosarcoma cells exposed to carcinogens. Although expressed in many normal cells, MET is overexpressed in many human cancers. MET is involved in the initiation and development of various human cancers and mediates proliferation, migration and invasion. Accordingly, MET has been successfully used as a biomarker for diagnosis and prognosis, survival, post-operative recurrence, risk assessment and pathologic grading, as well as a therapeutic target. In addition, recent work indicates that inhibition of MET expression and function has potential clinical benefit. This review summarizes the role, mechanism, and clinical significance of MET in the formation and development of human cancer.
Collapse
Affiliation(s)
- Xin Yang
- The Second Clinical Medical College, Lanzhou University, Lanzhou 730000, PR China; Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou 730000, PR China
| | - Hai-Yang Liao
- The Second Clinical Medical College, Lanzhou University, Lanzhou 730000, PR China; Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou 730000, PR China
| | - Hai-Hong Zhang
- The Second Clinical Medical College, Lanzhou University, Lanzhou 730000, PR China; Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou 730000, PR China.
| |
Collapse
|
36
|
Yan J, Zhao W, Yu W, Cheng H, Zhu B. LRRK2 correlates with macrophage infiltration in pan-cancer. Genomics 2021; 114:316-327. [PMID: 34929286 DOI: 10.1016/j.ygeno.2021.11.037] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 11/11/2021] [Accepted: 11/11/2021] [Indexed: 01/08/2023]
Abstract
Leucine-rich repeat kinase2 (LRRK2) influences the host immune responses and correlates with the pathogenesis of inflammation, cancer as well as Parkinson' Disease. Herein, we explored the oncogenic role of LRRK2 at pan-cancer level and validated the analysis by single cell RNA-sequencing and in-vitro experiments. As a result, LRRK2 significantly correlated with the survival events. Specifically, LRRK2 increased the risk of Low-Grade Glioma whereas improved the survival probability of patients with Skin Cutaneous Melanoma. Gene set enrichment analysis demonstrated the involvement of LRRK2 in the host immune responses. Within the tumor microenvironment, LRRK2 was positively associated with the recruitment of macrophages. Furthermore, scRNA-seq and co-culture experiments demonstrated that LRRK2 deficiency impaired macrophage functions, and influenced the neoplastic progression in a cancer type-specific manner. Therefore, the present study provided a therapeutic strategy for LGG based on the interference with LRRK2 expression and activity to prevent macrophage recruitment and promote tumor eradication.
Collapse
Affiliation(s)
- Jing Yan
- Department of Physiology, Jining Medical University, Jining City, Shandong Province 272067, China.
| | - Wenhui Zhao
- Department of Basic Medicine, Jiangsu College of Nursing, China
| | - Wei Yu
- Department of Physiology, Jining Medical University, Jining City, Shandong Province 272067, China
| | - Hongju Cheng
- Department of Physiology, Jining Medical University, Jining City, Shandong Province 272067, China
| | - Baoliang Zhu
- Department of Physiology, Jining Medical University, Jining City, Shandong Province 272067, China
| |
Collapse
|
37
|
Meza L, Malhotra J, Favorito C, Pal SK. Cabozantinib plus immunotherapy combinations in metastatic renal cell and urothelial carcinoma. Future Oncol 2021; 18:21-33. [PMID: 34766841 DOI: 10.2217/fon-2021-0570] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Treatment options for metastatic renal cell carcinoma (mRCC) and metastatic urothelial carcinoma (mUC) have increased dramatically over the past decade. However, even when novel approaches have proven to be effective as monotherapy, many patients still develop progressive disease, and different strategies are needed to increase clinical response and quality of life. Strategies combining targeted therapy (TT) and immunotherapy (IO) have emerged as a way to shorten the gap between responders and nonresponders to monotherapy and have reported promising results. In this review, we discuss the current role of cabozantinib in combination with IO agents in the treatment of metastatic RCC and UC and go over future directions in the field.
Collapse
Affiliation(s)
- Luis Meza
- Department of Medical Oncology & Experimental Therapeutics, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Jasnoor Malhotra
- Department of Medical Oncology & Experimental Therapeutics, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | | | - Sumanta K Pal
- Department of Medical Oncology & Experimental Therapeutics, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| |
Collapse
|
38
|
Cancel M, Fromont G, Blonz C, Chevreau C, Rioux-Leclercq N, Laguerre B, Oudard S, Gross-Goupil M, Gravis G, Goldwasser F, Rolland F, Delva R, Moise L, Emambux S, Vassal C, Zanetta S, Penel N, Fléchon A, Barthélémy P, Saldana C, Lefort F, Escudier B, Linassier C, Albiges L. Everolimus or sunitinib as first-line treatment of metastatic papillary renal cell carcinoma: A retrospective study of the GETUG group (Groupe d'Etude des Tumeurs Uro-Génitales). Eur J Cancer 2021; 158:1-11. [PMID: 34619467 DOI: 10.1016/j.ejca.2021.08.046] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 08/24/2021] [Accepted: 08/31/2021] [Indexed: 11/30/2022]
Abstract
BACKGROUND Two phase II trials (NCT00688753 and NCT00541008) reported efficacy data of sunitinib and everolimus in first-line treatment of metastatic papillary renal cell carcinoma (mpRCC). Although most patients receive sunitinib or a mammalian target of rapamycin (mTOR) inhibitor in first- and second-line treatment, the optimal strategy remained unknown. MATERIAL AND METHODS In 23 centres of the Groupe d'Etude des Tumeurs Urogénitales group, after centralised pathological review, we analysed retrospectively progression-free survival (PFS) of patients with mpRCC treated in first-line treatment (PFS-1) with sunitinib or everolimus (primary end-point), PFS in second-line treatment (PFS-2), overall survival (OS), objective response rate, disease control rate (DCR), overall sequence and prognostic factors for OS (secondary end-points). RESULTS One hundred thirty-eight patients (119 men and 19 women), median age 62.5 years, with mpRCC type 1 (n = 24) or non-type 1 (n = 114), received first-line sunitinib (n = 107) or everolimus (n = 31). With a median follow-up of 92 months, we found no significant difference between the treatment groups in terms of PFS-1 (5.5 versus 6.2 months) and DCR (69% versus 83%). Ninety-eight patients received a second-line treatment, 69% with mTOR inhibitors after sunitinib and 100% with tyrosine kinase inhibitors after everolimus, with similar DCR (64% versus 58%), median PFS-2 (3.4 versus 4.8 months) and OS (16.0 versus 20.3 months). No factor was prognostic for PFS-1, whereas leukocytosis, anaemia and the time from diagnosis to first systemic therapy < 1 year were prognostic for OS. We found no prognostic difference between both pRCC subtypes. The International Metastatic Renal Cell Database Consortium risk factors were prognostic for OS. CONCLUSION Sunitinib and everolimus had similar efficacy in first-line treatment of patients with mpRCC.
Collapse
Affiliation(s)
- Mathilde Cancel
- Department of Medical Oncology, CHU Bretonneau Tours, France
| | | | - Cyriac Blonz
- Department of Medical Oncology, Institut de Cancérologie de L'Ouest, Saint-Herblain, France
| | - Christine Chevreau
- Department of Medical Oncology, IUCT Oncopole - CLCC Institut Claudius Regaud, Toulouse, France
| | | | | | - Stéphane Oudard
- Department of Medical Oncology, CHU Hôpital Européen Georges Pompidou, Paris, France
| | | | - Gwenaelle Gravis
- Department of Medical Oncology, Institut Paoli-Calmettes, Marseille, France
| | | | - Frédéric Rolland
- Department of Medical Oncology, Institut de Cancérologie de L'Ouest, Saint-Herblain, France
| | - Rémy Delva
- Department of Medical Oncology, Institut de Cancérologie de L'Ouest, Angers, France
| | - Laura Moise
- Department of Medical Oncology, Centre François Baclesse, Caen, France
| | - Sheik Emambux
- Department of Medical Oncology, CHU La Milétrie, Poitiers, France
| | - Cécile Vassal
- Department of Medical Oncology, Institut de Cancérologie Lucien Neuwirth, Saint-Priest-en-Jarez, France
| | - Sylvie Zanetta
- Department of Medical Oncology, Centre Georges-François Leclerc, Dijon, France
| | - Nicolas Penel
- Lille University and Department of Medical Oncology, Centre Oscar Lambret, Lille, France
| | - Aude Fléchon
- Department of Medical Oncology, Centre Léon Bérard, Lyon, France
| | - Philippe Barthélémy
- Department of Medical Oncology, Institut de Cancérologie Strasbourg Europe, Strasbourg, France
| | | | - Félix Lefort
- Department of Medical Oncology, CHU Saint-André, Bordeaux, France
| | - Bernard Escudier
- Department of Medical Oncology, Gustave Roussy, Villejuif, France
| | | | - Laurence Albiges
- Department of Medical Oncology, Gustave Roussy, Villejuif, France
| |
Collapse
|
39
|
Halabi S, Yang Q, Carmack A, Zhang S, Foo WC, Eisen T, Stadler WM, Jones RJ, Garcia JA, Vaishampayan UN, Picus J, Hawkins RE, Hainsworth JD, Kollmannsberger CK, Logan TF, Puzanov I, Pickering LM, Ryan CW, Protheroe A, George DJ, Armstrong AJ. Tissue based biomarkers in non-clear cell RCC: Correlative analysis from the ASPEN clinical trial. KIDNEY CANCER JOURNAL : OFFICIAL JOURNAL OF THE KIDNEY CANCER ASSOCIATION 2021; 19:64-72. [PMID: 34765076 PMCID: PMC8580377 DOI: 10.52733/kcj19n3-a1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Biomarkers are needed in patients with non-clear cell renal cell carcinomas (NC-RCC), particularly papillary renal cell carcinoma, in order to inform on initial treatment selection and identify potentially novel targets for therapy. We enrolled 108 patients in ASPEN, an international randomized open-label phase 2 trial of patients with metastatic papillary, chromophobe, or unclassified NC-RCC treated with the mTOR inhibitor everolimus (n=57) or the vascular endothelial growth factor (VEGF) receptor inhibitor sunitinib (n=51), stratified by MSKCC risk and histology. The primary endpoint was overall survival (OS) and secondary efficacy endpoints for this exploratory biomarker analysis were radiographic progression-free survival (rPFS) defined by intention-to-treat using the RECIST 1.1 criteria and radiographic response rates. Tissue biomarkers (n=78) of mTOR pathway activation (phospho-S6 and -Akt, c-kit) and VEGF pathway activation (HIF-1α, c-MET) were prospectively explored in tumor tissue by immunohistochemistry prior to treatment and associated with clinical outcomes. We found that S6 activation was more common in poor risk NC-RCC tumors and S6/Akt activation was associated with worse PFS and OS outcomes with both everolimus and sunitinib, while c-kit was commonly expressed in chromophobe tumors and associated with improved outcomes with both agents. C-MET was commonly expressed in papillary tumors and was associated with lower rates of radiographic response but did not predict PFS for either agent. In multivariable analysis, both pAkt and c-kit were statistically significant prognostic biomarkers of OS. No predictive biomarkers of treatment response were identified for clinical outcomes. Most biomarker subgroups had improved outcomes with sunitinib as compared to everolimus.
Collapse
Affiliation(s)
- Susan Halabi
- Department of Biostatistics and Bioinformatics, Duke University, Durham NC
| | - Qian Yang
- Department of Biostatistics and Bioinformatics, Duke University, Durham NC
| | - Andrea Carmack
- Department of Biostatistics and Bioinformatics, Duke University, Durham NC
| | - Shiqi Zhang
- Department of Biostatistics and Bioinformatics, Duke University, Durham NC
| | - Wen-Chi Foo
- Department of Biostatistics and Bioinformatics, Duke University, Durham NC
| | - Tim Eisen
- University of Cambridge, Cambridge, United Kingdom
| | | | - Robert J. Jones
- University of Glasgow, The Beatson West of Scotland Cancer Centre, Glasgow, United Kingdom
| | | | - Ulka N. Vaishampayan
- University of Michigan, Ann Arbor/Karmanos Cancer Institute, Wayne State University, Detroit, MI USA
| | - Joel Picus
- Washington University in St. Louis, St. Louis, MO USA
| | | | | | | | - Theodore F. Logan
- Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN USA
| | - Igor Puzanov
- Vanderbilt University Medical Center, Nashville, TN USA
| | | | - Christopher W. Ryan
- Oregon Health & Science University, OHSU Knight Cancer Institute, Portland, OR USA
| | - Andrew Protheroe
- University of Oxford Medical Oncology Department, Oxford, United Kingdom
| | - Daniel J. George
- Duke University and the Duke Cancer Institute Center for Prostate and Urologic Cancers, Durham, NC
| | - Andrew J. Armstrong
- Duke University and the Duke Cancer Institute Center for Prostate and Urologic Cancers, Durham, NC
| |
Collapse
|
40
|
Vitale MG, Nasso C, Oltrecolli M, Baldessari C, Fanelli M, Dominici M, Sabbatini R. Cabozantinib and nivolumab as first-line treatment in advanced renal cell carcinoma. Expert Rev Anticancer Ther 2021; 21:1183-1192. [PMID: 34424125 DOI: 10.1080/14737140.2021.1971519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION In the last decade, there have been substantial changes in the management of metastatic renal cell carcinoma (mRCC) with combined regimens with immune checkpoint inhibitors (ICI) replacing targeted therapies. These combined regimens include the combination of cabozantinib plus nivolumab. AREAS COVERED Here, we provide an overview of clinical trials evaluating the combination of cabozantinib and nivolumab and the current clinical data on mechanism of action, pharmacokinetics, efficacy, and safety profile. EXPERT OPINION Dual immune checkpoint inhibition with nivolumab and ipilimumab as well as the combination of a vascular endothelial growth factor (VEGF) inhibitor and an immune checkpoint inhibitor have shown to improve outcomes in phase III trials in comparison to sunitinib (axitinib plus pembrolizumab, axitinib plus avelumab, bevacizumab plus atezolizumab, cabozantinib plus nivolumab, lenvatinib plus pembrolizumab). However, to date, there are no head-to-head trials comparing these new combination therapies and no biomarkers are available to guide the optimal choice of first line therapy.
Collapse
Affiliation(s)
| | - Cecilia Nasso
- Department of Oncology and Haematology, Modena University Hospital, Modena, Italy
| | - Marco Oltrecolli
- Department of Oncology and Haematology, Modena University Hospital, Modena, Italy
| | - Cinzia Baldessari
- Department of Oncology and Haematology, Modena University Hospital, Modena, Italy
| | - Martina Fanelli
- Department of Oncology and Haematology, Modena University Hospital, Modena, Italy
| | - Massimo Dominici
- Department of Oncology and Haematology, Modena University Hospital, Modena, Italy
| | - Roberto Sabbatini
- Department of Oncology and Haematology, Modena University Hospital, Modena, Italy
| |
Collapse
|
41
|
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: 16] [Impact Index Per Article: 4.0] [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
|
42
|
Lee M, Jain P, Wang F, Ma PC, Borczuk A, Halmos B. MET alterations and their impact on the future of non-small cell lung cancer (NSCLC) targeted therapies. Expert Opin Ther Targets 2021; 25:249-268. [PMID: 33945380 DOI: 10.1080/14728222.2021.1925648] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Introduction: The MET gene and its pathway normally plays a crucial role in cell homeostasis, motility, and apoptosis. However, when the MET gene is altered, there is an imbalance toward cell proliferation and invasion commonly seen in numerous different types of cancers. The heterogeneous group of MET alterations that includes MET amplification, MET exon 14 skipping mutation, and MET fusions has been difficult to diagnose and treat. Currently, treatments are focused on tyrosine kinase inhibitors but now there is emerging data on novel MET-targeted therapies including monoclonal antibodies and antibody-drug conjugates that have emerged.Areas covered: We introduce new emerging data on MET alterations in non-small cell lung cancer (NSCLC) that has contributed to advances in MET targeted therapeutics. We offer our perspective and examine new information on the mechanisms of the MET alterations in this review.Expert opinion: Given the trends currently involving the targeting of MET altered malignancies, there will most likely be a continued rapid expansion of testing, novel tyrosine kinase inhibitors and potent antibody approaches. Combination treatments will be necessary to optimize management of advanced and early disease.
Collapse
Affiliation(s)
- Matthew Lee
- Department of Oncology, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, NY, USA
| | - Prantesh Jain
- Division of Medical Oncology, Department of Medicine, University Hospitals Cleveland Medical Center, Case Comprehensive Cancer Center, Cleveland, OH, USA
| | - Feng Wang
- Department of Oncology, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, NY, USA
| | - Patrick C Ma
- Penn State CancerInstitute, PennState College of Medicine, Penn State Health Milton S Hershey Medical Center, Hershey, PA, USA
| | - Alain Borczuk
- Department of Pathology, NewYork-Presbyterian Hospital/Weill Cornell Medical Center, New York, NY, USA
| | - Balazs Halmos
- Department of Oncology, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
43
|
Marchetti A, Rosellini M, Mollica V, Rizzo A, Tassinari E, Nuvola G, Cimadamore A, Santoni M, Fiorentino M, Montironi R, Massari F. The Molecular Characteristics of Non-Clear Cell Renal Cell Carcinoma: What's the Story Morning Glory? Int J Mol Sci 2021; 22:6237. [PMID: 34207825 PMCID: PMC8226484 DOI: 10.3390/ijms22126237] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 06/06/2021] [Accepted: 06/08/2021] [Indexed: 01/05/2023] Open
Abstract
Non-clear cell renal cell carcinomas are a miscellaneous group of tumors that include different histological subtypes, each one characterized by peculiarity in terms of genetic alteration, clinical behavior, prognosis, and treatment response. Because of their low incidence and poor enrollment in clinical trials, alongside their heterogeneity, additional efforts are required to better unveil the pathogenetic mechanisms and, consequently, to improve the treatment algorithm. Nowadays, tyrosine kinase inhibitors, mTOR and MET inhibitors, and even cisplatin-based chemotherapy and immunotherapy are potential weapons that are still under evaluation in this setting. Various biomarkers have been evaluated for detecting progression and monitoring renal cell carcinoma, but more studies are necessary to improve this field. In this review, we provide an overview on the molecular characteristics of this group of tumors and the recently published trials, giving an insight into what might become the future therapeutic standard in this complex world of non-clear cell kidney cancers.
Collapse
Affiliation(s)
- Andrea Marchetti
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni—15, 40138 Bologna, Italy; (A.M.); (M.R.); (V.M.); (A.R.); (E.T.); (G.N.)
| | - Matteo Rosellini
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni—15, 40138 Bologna, Italy; (A.M.); (M.R.); (V.M.); (A.R.); (E.T.); (G.N.)
| | - Veronica Mollica
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni—15, 40138 Bologna, Italy; (A.M.); (M.R.); (V.M.); (A.R.); (E.T.); (G.N.)
| | - Alessandro Rizzo
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni—15, 40138 Bologna, Italy; (A.M.); (M.R.); (V.M.); (A.R.); (E.T.); (G.N.)
| | - Elisa Tassinari
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni—15, 40138 Bologna, Italy; (A.M.); (M.R.); (V.M.); (A.R.); (E.T.); (G.N.)
| | - Giacomo Nuvola
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni—15, 40138 Bologna, Italy; (A.M.); (M.R.); (V.M.); (A.R.); (E.T.); (G.N.)
| | - Alessia Cimadamore
- Section of Pathological Anatomy, School of Medicine, Polytechnic University of the Marche Region, United Hospitals, 60126 Ancona, Italy; (A.C.); (R.M.)
| | - Matteo Santoni
- Oncology Unit, Macerata Hospital, 62100 Macerata, Italy;
| | - Michelangelo Fiorentino
- Department of Specialistic Diagnostic and Experimental Medicine, University of Bologna, Via Massarenti 9, 40138 Bologna, Italy;
| | - Rodolfo Montironi
- Section of Pathological Anatomy, School of Medicine, Polytechnic University of the Marche Region, United Hospitals, 60126 Ancona, Italy; (A.C.); (R.M.)
| | - Francesco Massari
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni—15, 40138 Bologna, Italy; (A.M.); (M.R.); (V.M.); (A.R.); (E.T.); (G.N.)
| |
Collapse
|
44
|
de Vries-Brilland M, McDermott DF, Suárez C, Powles T, Gross-Goupil M, Ravaud A, Flippot R, Escudier B, Albigès L. Checkpoint inhibitors in metastatic papillary renal cell carcinoma. Cancer Treat Rev 2021; 99:102228. [PMID: 34111642 DOI: 10.1016/j.ctrv.2021.102228] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 05/13/2021] [Accepted: 05/17/2021] [Indexed: 02/02/2023]
Abstract
Papillary Renal Cell Carcinoma (pRCC) is the most common non-clear cell RCC (nccRCC) and a distinct entity, although heterogenous, associated with poor outcomes. The treatment landscape of metastatic pRCC (mpRCC) relied so far on targeted therapies, mimicking previous developments in metastatic clear-cell renal cell carcinoma. However, antiangiogenics as well as mTOR inhibitors retain only limited activity in mpRCC. As development of immune checkpoint inhibitors (ICI) is now underway in patients with mpRCC, we aimed at discussing early activity data and potential for future therapeutic strategies in monotherapy or combination. Expression of immune checkpoints such as PD-L1 and infiltrative immune cells in pRCC could provide insights into their potential immunogenicity, although this is currently poorly described. Based on retrospective and prospective data, efficacy of ICI as single agent remains limited. Combinations with tyrosine-kinase inhibitors, notably with anti-MET inhibitors, harbor promising response rates and may enter the standard of care in untreated patients. Collaborative work is needed to refine the molecular and immune landscape of pRCC, and pursue efforts to set up predictive biomarker-driven clinical trials in these rare tumors.
Collapse
Affiliation(s)
- M de Vries-Brilland
- Department of Medical Oncology, Integrated Centers of Oncology (ICO) Paul Papin, Angers, France
| | - D F McDermott
- Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - C Suárez
- Medical Oncology, Vall d́Hebron Institute of Oncology (VHIO), Hospital Universitari Vall d́Hebron, Vall d́Hebron Barcelona Hospital Campus, Spain
| | - T Powles
- The Royal Free NHS Trust and Barts Cancer Institute, Queen Mary University of London, London, UK
| | - M Gross-Goupil
- Department of Medical Oncology, Hôpital Saint-André, University of Bordeaux-CHU, Bordeaux, France
| | - A Ravaud
- Department of Medical Oncology, Hôpital Saint-André, University of Bordeaux-CHU, Bordeaux, France
| | - R Flippot
- Medical Oncology, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - B Escudier
- Medical Oncology, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - L Albigès
- Medical Oncology, Gustave Roussy, Université Paris-Saclay, Villejuif, France.
| |
Collapse
|
45
|
Patel HV, Srivastava A, Srinivasan R, Singer EA. A challenging frontier - the genomics and therapeutics of nonclear cell renal cell carcinoma. Curr Opin Oncol 2021; 33:212-220. [PMID: 33818540 PMCID: PMC8244822 DOI: 10.1097/cco.0000000000000721] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW As molecular profiling of renal cell carcinoma (RCC) continues to elucidate novel targets for nonclear cell histologies, understanding the landscape of these targets is of utmost importance. In this review, we highlight the genomic landscape of nonclear cell RCC and its implications for current and future systemic therapies. RECENT FINDINGS Several genomic studies have described the mutational burden among nonclear cell histologies. These studies have highlighted the importance of MET in papillary RCC and led to several clinical trials evaluating the efficacy of MET inhibitors for papillary RCC. The success of immune checkpoint inhibitors, such as ipilimumab and nivolumab, in clear cell RCC has led to ongoing trials evaluating these novel therapeutics in nonclear cell RCC. SUMMARY Genomic profiling has allowed for the evaluation of novel targets for nonclear cell RCC. This evolving therapeutic landscape is being explored in promising, ongoing trials that have the potential for changing how nonclear cell RCC is managed.
Collapse
Affiliation(s)
- Hiren V. Patel
- Section of Urologic Oncology, Rutgers Cancer Institute of New Jersey and Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey, USA
| | - Arnav Srivastava
- Section of Urologic Oncology, Rutgers Cancer Institute of New Jersey and Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey, USA
| | - Ramaprasad Srinivasan
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Eric A. Singer
- Section of Urologic Oncology, Rutgers Cancer Institute of New Jersey and Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey, USA
| |
Collapse
|
46
|
Yao HP, Tong XM, Wang MH. Oncogenic mechanism-based pharmaceutical validation of therapeutics targeting MET receptor tyrosine kinase. Ther Adv Med Oncol 2021; 13:17588359211006957. [PMID: 33868463 PMCID: PMC8020248 DOI: 10.1177/17588359211006957] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Accepted: 03/11/2021] [Indexed: 02/06/2023] Open
Abstract
Aberrant expression and/or activation of the MET receptor tyrosine kinase is
characterized by genomic recombination, gene amplification, activating mutation,
alternative exon-splicing, increased transcription, and their different
combinations. These dysregulations serve as oncogenic determinants contributing
to cancerous initiation, progression, malignancy, and stemness. Moreover,
integration of the MET pathway into the cellular signaling network as an
addiction mechanism for survival has made this receptor an attractive
pharmaceutical target for oncological intervention. For the last 20 years,
MET-targeting small-molecule kinase inhibitors (SMKIs), conventional therapeutic
monoclonal antibodies (TMABs), and antibody-based biotherapeutics such as
bispecific antibodies, antibody–drug conjugates (ADC), and dual-targeting ADCs
have been under intensive investigation. Outcomes from preclinical studies and
clinical trials are mixed with certain successes but also various setbacks. Due
to the complex nature of MET dysregulation with multiple facets and underlying
mechanisms, mechanism-based validation of MET-targeting therapeutics is crucial
for the selection and validation of lead candidates for clinical trials. In this
review, we discuss the importance of various types of mechanism-based
pharmaceutical models in evaluation of different types of MET-targeting
therapeutics. The advantages and disadvantages of these mechanism-based
strategies for SMKIs, conventional TMABs, and antibody-based biotherapeutics are
analyzed. The demand for establishing new strategies suitable for validating
novel biotherapeutics is also discussed. The information summarized should
provide a pharmaceutical guideline for selection and validation of MET-targeting
therapeutics for clinical application in the future.
Collapse
Affiliation(s)
- Hang-Ping Yao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiang-Min Tong
- Department of Hematology, Zhejiang Provincial People's Hospital and People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Ming-Hai Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, First Affiliated hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
47
|
MET alterations in biphasic squamoid alveolar papillary renal cell carcinomas and clinicopathological features. Mod Pathol 2021; 34:647-659. [PMID: 32770124 DOI: 10.1038/s41379-020-0645-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 07/24/2020] [Accepted: 07/25/2020] [Indexed: 11/09/2022]
Abstract
Biphasic squamoid alveolar papillary renal cell carcinoma (BSA-PRCC) is a recently studied lesion considered a morphologic variant of papillary renal cell carcinoma (RCC), more closely related to type 1. Considering the role of proto-oncogene MET in both sporadic type 1 papillary RCC and hereditary papillary RCC, we aimed to explore the role of MET activation in the oncogenesis of BSA-PRCC. We identified 17 patients with either unique (n = 14) or multiple (n = 3) BSA-PRCC, all localized, and performed an integrative analysis of MET status in 18 formalin-fixed paraffin-embedded tumors combining next-generation sequencing analysis, fluorescent in situ hybridization and immunohistochemistry. Trisomy 7 was found in 86% of tumors (14/16) without MET amplification at 7q31 (15/15). A pathogenic MET genetic variant was identified in 60% (9/15) of cases, at the germline level in 57% (4/7) of tested patients or at the somatic level (5/11). MET expression was observed in all tumors with a higher value of combined score in large cells (mean 97%, range 80-100%) than in small cells (mean 74%, range 10-100%) and was lower in two cases without MET copy number gain. In conclusion, our study provides additional evidence to consider biphasic squamoid alveolar papillary RCC as a morphological variant of type 1 papillary renal RCC. Our data strongly suggest that MET represents a major oncogenic driver gene in BSA-PRCC, harboring a higher frequency of MET mutation that encourages to further explore the benefice of anti-MET targeted therapies for aggressive BSA-PRCC.
Collapse
|
48
|
Pal SK, Tangen C, Thompson IM, Balzer-Haas N, George DJ, Heng DYC, Shuch B, Stein M, Tretiakova M, Humphrey P, Adeniran A, Narayan V, Bjarnason GA, Vaishampayan U, Alva A, Zhang T, Cole S, Plets M, Wright J, Lara PN. A comparison of sunitinib with cabozantinib, crizotinib, and savolitinib for treatment of advanced papillary renal cell carcinoma: a randomised, open-label, phase 2 trial. Lancet 2021; 397:695-703. [PMID: 33592176 PMCID: PMC8687736 DOI: 10.1016/s0140-6736(21)00152-5] [Citation(s) in RCA: 160] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/14/2021] [Accepted: 01/20/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND MET (also known as hepatocyte growth factor receptor) signalling is a key driver of papillary renal cell carcinoma (PRCC). Given that no optimal therapy for metastatic PRCC exists, we aimed to compare an existing standard of care, sunitinib, with the MET kinase inhibitors cabozantinib, crizotinib, and savolitinib for treatment of patients with PRCC. METHODS We did a randomised, open-label, phase 2 trial done in 65 centres in the USA and Canada. Eligible patients were aged 18 years or older with metastatic PRCC who had received up to one previous therapy (excluding vascular endothelial growth factor-directed and MET-directed agents). Patients were randomly assigned to receive sunitinib, cabozantinib, crizotinib, or savolitinib, with stratification by receipt of previous therapy and PRCC subtype. All drug doses were administered orally: sunitinib 50 mg, 4 weeks on and 2 weeks off (dose reductions to 37·5 mg and 25 mg allowed); cabozantinib 60 mg daily (reductions to 40 mg and 20 mg allowed); crizotinib 250 mg twice daily (reductions to 200 mg twice daily and 250 mg once daily allowed); and savolitinib 600 mg daily (reductions to 400 mg and 200 mg allowed). Progression-free survival (PFS) was the primary endpoint. Analyses were done in an intention-to-treat population, with patients who did not receive protocol therapy excluded from safety analyses. This trial is registered with ClinicalTrials.gov, NCT02761057. FINDINGS Between April 5, 2016, and Dec 15, 2019, 152 patients were randomly assigned to one of four study groups. Five patients were identified as ineligible post-randomisation and were excluded from these analyses, resulting in 147 eligible patients. Assignment to the savolitinib (29 patients) and crizotinib (28 patients) groups was halted after a prespecified futility analysis; planned accrual was completed for both sunitinib (46 patients) and cabozantinib (44 patients) groups. PFS was longer in patients in the cabozantinib group (median 9·0 months, 95% CI 6-12) than in the sunitinib group (5·6 months, 3-7; hazard ratio for progression or death 0·60, 0·37-0·97, one-sided p=0·019). Response rate for cabozantinib was 23% versus 4% for sunitinib (two-sided p=0·010). Savolitinib and crizotinib did not improve PFS compared with sunitinib. Grade 3 or 4 adverse events occurred in 31 (69%) of 45 patients receiving sunitinib, 32 (74%) of 43 receiving cabozantinib, ten (37%) of 27 receiving crizotinib, and 11 (39%) of 28 receiving savolitinib; one grade 5 thromboembolic event was recorded in the cabozantinib group. INTERPRETATION Cabozantinib treatment resulted in significantly longer PFS compared with sunitinib in patients with metastatic PRCC. FUNDING National Institutes of Health and National Cancer Institute.
Collapse
Affiliation(s)
- Sumanta K Pal
- City of Hope Comprehensive Cancer Center, Duarte, CA, USA.
| | - Catherine Tangen
- SWOG Statistics and Data Management Center, Seattle, WA, USA; Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Ian M Thompson
- CHRISTUS Santa Rosa Medical Center Hospital, San Antonio, TX, USA
| | - Naomi Balzer-Haas
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Daniel J George
- Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | | | - Brian Shuch
- Institute of Urologic Oncology, University of California Los Angeles, Los Angeles, CA, USA
| | - Mark Stein
- Department of Medicine, Columbia University, New York, NY, USA
| | - Maria Tretiakova
- Department of Pathology, University of Washington, Seattle, WA, USA
| | - Peter Humphrey
- City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | | | - Vivek Narayan
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Ulka Vaishampayan
- Department of Medicine, Wayne State University, Detroit, MI, USA; Department of Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Ajjai Alva
- Department of Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Tian Zhang
- Alliance for Clinical Trials in Oncology, Duke Cancer Research Institute, Durham, NC, USA
| | - Scott Cole
- Oklahoma Cancer Specialists and Research Institute, NRG Oncology, Tulsa, OK, USA
| | - Melissa Plets
- City of Hope Comprehensive Cancer Center, Duarte, CA, USA; Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - John Wright
- Cancer Therapy Evaluation Program, Investigational Drug Branch, National Cancer Institute, Bethesda, MD, USA
| | - Primo N Lara
- University of California Davis Comprehensive Cancer Center, Sacramento, CA, USA
| |
Collapse
|
49
|
Borchiellini D, Barthélémy P. Cabozantinib: a new first-line option for papillary renal cell carcinoma? Lancet 2021; 397:645-647. [PMID: 33592177 DOI: 10.1016/s0140-6736(21)00316-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 02/03/2021] [Indexed: 12/30/2022]
|
50
|
Jones RDO, Grondine M, Borodovsky A, San Martin M, DuPont M, D'Cruz C, Schuller A, Henry R, Barry E, Castriotta L, Anjum R, Petersson K, Sahota T, Ahmed GF. A pharmacokinetic-pharmacodynamic model for the MET tyrosine kinase inhibitor, savolitinib, to explore target inhibition requirements for anti-tumour activity. Br J Pharmacol 2020; 178:600-613. [PMID: 33125717 DOI: 10.1111/bph.15301] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 10/05/2020] [Accepted: 10/20/2020] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND AND PURPOSE Savolitinib (AZD6094, HMPL-504, volitinib) is an oral, potent, and highly MET receptor TK inhibitor. This series of studies aimed to develop a pharmacokinetic-pharmacodynamic (PK/PD) model to link inhibition of MET phosphorylation (pMET) by savolitinib with anti-tumour activity. EXPERIMENTAL APPROACH Cell line-derived xenograft (CDX) experiments using human lung cancer (EBC-1) and gastric cancer (MKN-45) cells were conducted in athymic nude mice using a variety of doses and schedules of savolitinib. Tumour pMET changes and growth inhibition were calculated after 28 days. Population PK/PD techniques were used to construct a PK/PD model for savolitinib. KEY RESULTS Savolitinib showed dose- and dose frequency-dependent anti-tumour activity in the CDX models, with more frequent, lower dosing schedules (e.g., twice daily) being more effective than intermittent, higher dosing schedules (e.g., 4 days on/3 days off or 2 days on/5 days off). There was a clear exposure-response relationship, with maximal suppression of pMET of >90%. Data from additional CDX and patient-derived xenograft (PDX) models overlapped, allowing calculation of a single EC50 of 0.38 ng·ml-1 . Tumour growth modelling demonstrated that prolonged, high levels of pMET inhibition (>90%) were required for tumour stasis and regression in the models. CONCLUSION AND IMPLICATIONS High and persistent levels of MET inhibition by savolitinib were needed for optimal monotherapy anti-tumour activity in preclinical models. The modelling framework developed here can be used to translate tumour growth inhibition from the mouse to human and thus guide choice of clinical dose and schedule.
Collapse
Affiliation(s)
- Rhys D O Jones
- Oncology R&D, Research and Early Development, AstraZeneca, Cambridge, UK
| | - Mike Grondine
- Oncology R&D, Research and Early Development, AstraZeneca, Boston, Massachusetts, USA
| | - Alexandra Borodovsky
- Formerly Oncology R&D, Research and Early Development, AstraZeneca, Boston, Massachusetts, USA
| | - Maryann San Martin
- Oncology R&D, Research and Early Development, AstraZeneca, Boston, Massachusetts, USA
| | - Michelle DuPont
- Oncology R&D, Research and Early Development, AstraZeneca, Boston, Massachusetts, USA
| | - Celina D'Cruz
- Oncology R&D, Research and Early Development, AstraZeneca, Boston, Massachusetts, USA
| | - Alwin Schuller
- Oncology R&D, Research and Early Development, AstraZeneca, Boston, Massachusetts, USA
| | - Ryan Henry
- Formerly Oncology R&D, Research and Early Development, AstraZeneca, Boston, Massachusetts, USA
| | - Evan Barry
- Formerly Oncology R&D, Research and Early Development, AstraZeneca, Boston, Massachusetts, USA
| | - Lillian Castriotta
- Oncology R&D, Research and Early Development, AstraZeneca, Boston, Massachusetts, USA
| | - Rana Anjum
- Oncology R&D, Research and Early Development, AstraZeneca, Boston, Massachusetts, USA
| | | | - Tarjinder Sahota
- BioPharmaceuticals R&D, Clinical Pharmacology and Safety Sciences, AstraZeneca, Cambridge, UK
| | - Ghada F Ahmed
- Formerly BioPharmaceuticals R&D, Clinical Pharmacology and Safety Sciences, AstraZeneca, Cambridge, UK
| |
Collapse
|