1
|
Anti-angiogenesis and Immunotherapy: Novel Paradigms to Envision Tailored Approaches in Renal Cell-Carcinoma. J Clin Med 2020; 9:jcm9051594. [PMID: 32456352 PMCID: PMC7291047 DOI: 10.3390/jcm9051594] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 05/18/2020] [Accepted: 05/20/2020] [Indexed: 12/16/2022] Open
Abstract
Although decision making strategy based on clinico-histopathological criteria is well established, renal cell carcinoma (RCC) represents a spectrum of biological ecosystems characterized by distinct genetic and molecular alterations, diverse clinical courses and potential specific therapeutic vulnerabilities. Given the plethora of drugs available, the subtype-tailored treatment to RCC subtype holds the potential to improve patient outcome, shrinking treatment-related morbidity and cost. The emerging knowledge of the molecular taxonomy of RCC is evolving, whilst the antiangiogenic and immunotherapy landscape maintains and reinforces their potential. Although several prognostic factors of survival in patients with RCC have been described, no reliable predictive biomarkers of treatment individual sensitivity or resistance have been identified. In this review, we summarize the available evidence able to prompt more precise and individualized patient selection in well-designed clinical trials, covering the unmet need of medical choices in the era of next-generation anti-angiogenesis and immunotherapy.
Collapse
|
2
|
D'Aniello C, Berretta M, Cavaliere C, Rossetti S, Facchini BA, Iovane G, Mollo G, Capasso M, Pepa CD, Pesce L, D'Errico D, Buonerba C, Di Lorenzo G, Pisconti S, De Vita F, Facchini G. Biomarkers of Prognosis and Efficacy of Anti-angiogenic Therapy in Metastatic Clear Cell Renal Cancer. Front Oncol 2019; 9:1400. [PMID: 31921657 PMCID: PMC6917607 DOI: 10.3389/fonc.2019.01400] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Accepted: 11/27/2019] [Indexed: 12/30/2022] Open
Abstract
In the last decades, the prognosis of metastatic renal cell carcinoma (mRCC) has remarkably improved following the advent of the "targeted therapy" era. The expanding knowledge on the prominent role played by angiogenesis in RCC pathogenesis has led to approval of multiple anti-angiogenic agents such as sunitinib, pazopanib, axitinib, cabozantinib, sorafenib, and bevacizumab. These agents can induce radiological responses and delay cancer progression for months or years before onset of resistance, with a clinically meaningful activity. The need for markers of prognosis and efficacy of anti-angiogenic agents has become more compelling as novel systemic immunotherapy agents have also been approved in RCC and can be administered as an alternative to angiogenesis inhibitors. Anti PD-1 monoclonal antibody nivolumab has been approved in the second-line setting after tyrosine kinase inhibitors failure, while combination of nivolumab plus anti CTLA-4 monoclonal antibody ipilimumab has been approved as first-line therapy of RCC patients at intermediate or poor prognosis. In this review article, biomarkers of prognosis and efficacy of antiangiogenic therapies are summarized with a focus on those that have the potential to affect treatment decision-making in RCC. Biomarkers predictive of toxicity of anti-angiogenic agents have also been discussed.
Collapse
Affiliation(s)
- Carmine D'Aniello
- Division of Medical Oncology, A.O.R.N. dei COLLI “Ospedali Monaldi-Cotugno-CTO,”Naples, Italy
| | - Massimiliano Berretta
- Division of Medical Oncology, Istituto Nazionale Tumori, IRCCS CRO Aviano (PN), Milan, Italy
| | - Carla Cavaliere
- UOC of Medical Oncology, ASL NA 3 SUD, Ospedali Riuniti Area Nolana, Nola, Italy
| | - Sabrina Rossetti
- Departmental Unit of Experimental Uro-Andrologic Clinical Oncology, Istituto Nazionale Tumori Fondazione G. Pascale—IRCCS, Naples, Italy
| | - Bianca Arianna Facchini
- Division of Medical Oncology, Department of Precision Medicine, University of Campania 'Luigi Vanvitelli', Naples, Italy
| | - Gelsomina Iovane
- Departmental Unit of Experimental Uro-Andrologic Clinical Oncology, Istituto Nazionale Tumori Fondazione G. Pascale—IRCCS, Naples, Italy
| | - Giovanna Mollo
- Departmental Unit of Experimental Uro-Andrologic Clinical Oncology, Istituto Nazionale Tumori Fondazione G. Pascale—IRCCS, Naples, Italy
| | - Mariagrazia Capasso
- Departmental Unit of Experimental Uro-Andrologic Clinical Oncology, Istituto Nazionale Tumori Fondazione G. Pascale—IRCCS, Naples, Italy
| | | | - Laura Pesce
- Oncology Unit, San Luca Hospital, Vallo Della Lucania, Italy
| | - Davide D'Errico
- Departmental Unit of Experimental Uro-Andrologic Clinical Oncology, Istituto Nazionale Tumori Fondazione G. Pascale—IRCCS, Naples, Italy
| | - Carlo Buonerba
- CRTR Rare Tumors Reference Center, AOU Federico II, Naples, Italy
- Environment & Health Operational Unit, Zoo-Prophylactic Institute of Southern Italy, Portici, Italy
| | - Giuseppe Di Lorenzo
- Department of Clinical Medicine and Surgery, University Federico II, Naples, Italy
- Department of Medicine, University of Molise, Campobasso, Italy
| | - Salvatore Pisconti
- Department of Onco-Hematology, Medical Oncology, S.G. Moscati Hospital, Taranto, Italy
| | - Ferdinando De Vita
- Division of Medical Oncology, Department of Precision Medicine, University of Campania 'Luigi Vanvitelli', Naples, Italy
| | - Gaetano Facchini
- Departmental Unit of Experimental Uro-Andrologic Clinical Oncology, Istituto Nazionale Tumori Fondazione G. Pascale—IRCCS, Naples, Italy
| |
Collapse
|
3
|
The evolving role of cytoreductive nephrectomy: incorporating genomics of metastatic renal cell carcinoma into treatment decisions. Curr Opin Urol 2019; 29:531-539. [PMID: 31313716 DOI: 10.1097/mou.0000000000000663] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
PURPOSE OF REVIEW Recent publications evaluating cytoreductive nephrectomy in the era of targeted therapy emphasize the importance of patient selection. We reviewed the predictive role of genetic alterations in patients with metastatic renal cell carcinoma (mRCC) undergoing cytoreductive nephrectomy. RECENT FINDINGS Studies evaluating the association between genetic alterations and outcomes following systemic treatment for mRCC include mainly patients after cytoreductive nephrectomy. Expression of proangiogenic genes, single nucleotide polymorphisms involving genes of the vascular-endothelial growth factor (VEGF) pathway and somatic mutations of chromatin remodeling genes were associated with response to VEGF-targeted therapy. Outcomes following treatment with mammalian target of rapamycin (mTOR) inhibitors were initially associated with mTOR/TSC1/TSC2 mutations; however, subsequent studies did not validate these findings but rather found an association between loss of PTEN expression and PBRM1 mutations and improved outcomes. Loss of PBRM1 was initially linked to response to immunotherapy; however, larger studies question this association and showed high expression of T-effector gene signature predicted improved outcome. Primary tumors with low intratumor heterogeneity but elevated somatic copy-number alterations were associated with rapid progression at multiple sites. SUMMARY Genetic alterations may help select patients for cytoreductive nephrectomy and optimize timing of treatment. Intratumor heterogeneity and genetic discordance between primary and metastatic tumors may limit clinical applicability. Future studies should evaluate approaches to overcome these limitations.
Collapse
|
4
|
Vanmechelen M, Lambrechts D, Van Brussel T, Verbiest A, Couchy G, Schöffski P, Dumez H, Debruyne PR, Lerut E, Machiels JP, Richard V, Albersen M, Verschaeve V, Oudard S, Méjean A, Wolter P, Zucman-Rossi J, Beuselinck B. Fibroblast Growth Factor Receptor-2 Polymorphism rs2981582 is Correlated With Progression-free Survival and Overall Survival in Patients With Metastatic Clear-cell Renal Cell Carcinoma Treated With Sunitinib. Clin Genitourin Cancer 2019; 17:e235-e246. [DOI: 10.1016/j.clgc.2018.11.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 11/08/2018] [Accepted: 11/08/2018] [Indexed: 02/06/2023]
|
5
|
Liu X, Swen JJ, Diekstra MHM, Boven E, Castellano D, Gelderblom H, Mathijssen RHJ, Vermeulen SH, Oosterwijk E, Junker K, Roessler M, Alexiusdottir K, Sverrisdottir A, Radu MT, Ambert V, Eisen T, Warren A, Rodríguez-Antona C, García-Donas J, Böhringer S, Koudijs KKM, Kiemeney LALM, Rini BI, Guchelaar HJ. A Genetic Polymorphism in CTLA-4 Is Associated with Overall Survival in Sunitinib-Treated Patients with Clear Cell Metastatic Renal Cell Carcinoma. Clin Cancer Res 2018; 24:2350-2356. [PMID: 29490989 DOI: 10.1158/1078-0432.ccr-17-2815] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 01/22/2018] [Accepted: 02/22/2018] [Indexed: 11/16/2022]
Abstract
Purpose: The survival of patients with clear cell metastatic renal cell carcinoma (cc-mRCC) has improved substantially since the introduction of tyrosine kinase inhibitors (TKI). With the fact that TKIs interact with immune responses, we investigated whether polymorphisms of genes involved in immune checkpoints are related to the clinical outcome of cc-mRCC patients treated with sunitinib as first TKI.Experimental Design: Twenty-seven single-nucleotide polymorphisms (SNP) in CD274 (PD-L1), PDCD1 (PD-1), and CTLA-4 were tested for a possible association with progression-free survival (PFS) and overall survival (OS) in a discovery cohort of 550 sunitinib-treated cc-mRCC patients. SNPs with a significant association (P < 0.05) were tested in an independent validation cohort of 138 sunitinib-treated cc-mRCC patients. Finally, data of the discovery and validation cohort were pooled for meta-analysis.Results:CTLA-4 rs231775 and CD274 rs7866740 showed significant associations with OS in the discovery cohort after correction for age, gender, and Heng prognostic risk group [HR, 0.84; 95% confidence interval (CI), 0.72-0.98; P = 0.028, and HR, 0.73; 95% CI, 0.54-0.99; P = 0.047, respectively]. In the validation cohort, the associations of both SNPs with OS did not meet the significance threshold of P < 0.05. After meta-analysis, CTLA-4 rs231775 showed a significant association with OS (HR, 0.83; 95% CI, 0.72-0.95; P = 0.008). Patients with the GG genotype had longer OS (35.1 months) compared with patients with an AG (30.3 months) or AA genotype (24.3 months). No significant associations with PFS were found.Conclusions: The G-allele of rs231775 in the CTLA-4 gene is associated with an improved OS in sunitinib-treated cc-mRCC patients and could potentially be used as a prognostic biomarker. Clin Cancer Res; 24(10); 2350-6. ©2018 AACR.
Collapse
Affiliation(s)
- Xiaoyan Liu
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, the Netherlands.,Institute of Clinical Pharmacology, Qilu Hospital of Shandong University, Jinan, China
| | - Jesse J Swen
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, the Netherlands
| | - Meta H M Diekstra
- Department of Clinical Pharmacy and Toxicology, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Epie Boven
- Department of Medical Oncology, VU University Medical Center, Amsterdam, the Netherlands
| | - Daniel Castellano
- Oncology Department, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Hans Gelderblom
- Department of Medical Oncology, Leiden University Medical Center, Leiden, the Netherlands
| | - Ron H J Mathijssen
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Sita H Vermeulen
- Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Egbert Oosterwijk
- Department of Urology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Kerstin Junker
- Clinic of Urology and Paediatric Urology, Saarland University, Homburg, Germany
| | - Max Roessler
- CESAR Central Office, CESAR Central European Society for Anticancer Drug Research-EWIV, Vienna, Austria
| | | | | | - Marius T Radu
- University of Medicine and Pharmacy Carol Davila, Bucuresti, Romania
| | - Valentin Ambert
- University of Medicine and Pharmacy Carol Davila, Bucuresti, Romania
| | - Tim Eisen
- Department of Oncology, Cambridge University Hospitals NHS Foundation Trust, Addenbrooke's Hospital, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Anne Warren
- Department of Pathology, Cambridge University Hospitals NHS Foundation Trust, Addenbrooke's Hospital, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Cristina Rodríguez-Antona
- Hereditary Endocrine Cancer Group, Spanish National Cancer Research Centre (CNIO) and Biomedical Network on Rare Diseases (CIBERER), Madrid, Spain
| | - Jesus García-Donas
- Medical Oncology, HM Hospitales, Centro Integral Oncológico HM Clara Campal, Madrid, Spain
| | - Stefan Böhringer
- Department of Medical Statistics and Bioinformatics, Leiden University Medical Center, Leiden, the Netherlands
| | - Karel K M Koudijs
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, the Netherlands
| | - Lambertus A L M Kiemeney
- Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Brian I Rini
- Department of Solid Tumor Oncology, Cleveland Clinic Taussig Cancer Institute (CCF), Cleveland, Ohio
| | - Henk-Jan Guchelaar
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, the Netherlands.
| |
Collapse
|