1
|
Ciccarese C, Büttner T, Cerbone L, Zampiva I, Monteiro FSM, Basso U, Pichler M, Vitale MG, Fiala O, Roviello G, Kopp RM, Carrozza F, Pichler R, Grillone F, Calabuig EP, Zeppellini A, Küronya Z, Galli L, Facchini G, Sunela K, Mosca A, Molina-Cerrillo J, Spinelli GP, Ansari J, Scala A, Mollica V, Grande E, Buti S, Kanesvaran R, Zakopoulou R, Bamias A, Rizzo M, Massari F, Iacovelli R, Santoni M. Clinical features and response to immune combinations in patients with renal cell carcinoma and sarcomatoid de-differentiation (ARON-1 study). Int J Cancer 2024; 155:2036-2046. [PMID: 39243397 DOI: 10.1002/ijc.35141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 02/28/2024] [Accepted: 03/12/2024] [Indexed: 09/09/2024]
Abstract
Metastatic renal cell carcinoma (mRCC) carrying sarcomatoid features (sRCC) has aggressive biology and poor prognosis. First-line immunotherapy (IO)-based combinations have improved the outcome of clear cell RCC patients, including that of sRCC. Real-world data confirming the adequate first-line management of sRCC is largely lacking. We investigated the clinical features and the outcome of sRCC patients treated with IO-based combinations within the ARON-1 study population (NCT05287464). The primary objective was to define the incidence and baseline clinical characteristics of sRCC compared with non-sRCC patients. The secondary objective was to describe the outcome of sRCC patients based on type of first-line treatment (IO + IO vs. IO + tyrosin kinase inhibitor [TKI]). We identified 1362 mRCC patients with IMDC intermediate or poor risk, 226 sRCC and 1136 non-sRCC. These two subgroups did not differ in terms of baseline characteristics. The median overall survival (OS) was 26.8 months (95%CI 21.6-44.2) in sRCC and 35.3 months (95%CI 30.2-40.4) in non-sRCC patients (p = .013). The median progression-free survival (PFS) was longer in non-sRCC patients compared to sRCC (14.5 vs. 12.3 months, p = .064). In patients treated with first-line IO + TKI the median OS was 34.4 months compared to 26.4 months of those who received IO + IO (p = .729). The median PFS was 12.4 months with IO + TKI and 12.3 months with IO + IO (p = .606). In conclusion, we confirm that sRCC are aggressive tumors with poor prognosis. IO-based combinations improve survival outcomes of sRCC patients, regardless from the type of strategy (IO + IO versus IO + TKI) adopted.
Collapse
Affiliation(s)
- Chiara Ciccarese
- Medical Oncology Unit, Fondazione Policlinico A. Gemelli IRCCS, Rome, Italy
| | - Thomas Büttner
- Department of Urology, University Hospital Bonn (UKB), Bonn, Germany
| | - Linda Cerbone
- Department of Medical Oncology, San Camillo Forlanini Hospital, Rome, Italy
| | - Ilaria Zampiva
- Section of Innovation Biomedicine-Oncology Area, Department of Engineering for Innovation Medicine (DIMI), University of Verona and University and Hospital Trust (AOUI) of Verona, Verona, Italy
| | | | - Umberto Basso
- Oncology 3 Unit, Department of Oncology, Istituto Oncologico Veneto IOV IRCCS, Padova, Italy
| | - Martin Pichler
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Maria Giuseppa Vitale
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, Modena, Italy
| | - Ondrej Fiala
- Department of Oncology and Radiotherapeutics, Faculty of Medicine and University Hospital in Pilsen and Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| | - Giandomenico Roviello
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| | - Ray Manneh Kopp
- Clinical Oncology, Sociedad de oncología y hematología del Cesar, Valledupar, Colombia
| | - Francesco Carrozza
- Department of Medical Oncology, AUSL della Romagna, Ospedale Civile degli Infermi, Faenza, Italy
| | - Renate Pichler
- Department of Urology, Medical University of Innsbruck, Innsbruck, Austria
| | - Francesco Grillone
- Oncologia, Oncologia PO Pugliese Ciaccio Azienda Ospedaliera Universitaria Renato Dulbecco, Catanzaro, Italy
| | - Esther Pérez Calabuig
- Medical Oncology Department, CHU Insular-Materno Infantil, Las Palmas de Gran Canaria, Spain
| | - Annalisa Zeppellini
- Medical Oncology, Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Zsófia Küronya
- Department of Genitourinary Medical Oncology and Clinical Pharmacology, National Institute of Oncology, Budapest, Hungary
| | - Luca Galli
- Oncology Unit 2, University Hospital of Pisa, Pisa, Italy
| | - Gaetano Facchini
- Oncology Operative Unit, "Santa Maria delle Grazie" Hospital, Pozzuoli, Italy
| | - Kaisa Sunela
- Department of Oncology, Tampere University Hospital, Tampere Cancer Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Alessandra Mosca
- Oncology Department, Candiolo Cancer Institute, IRCCS-FPO, Torino, Italy
| | | | | | | | - Alessandro Scala
- Medical Oncology Unit, Fondazione Policlinico A. Gemelli IRCCS, Rome, Italy
| | - Veronica Mollica
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Enrique Grande
- Department of Medical Oncology, MD Anderson Cancer Center Madrid, Madrid, Spain
| | - Sebastiano Buti
- Medical Oncology Unit, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | | | - Roubini Zakopoulou
- 2nd Propaedeutic Department of Internal Medicine, School of Medicine, ATTIKON University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Aristotelis Bamias
- 2nd Propaedeutic Department of Internal Medicine, School of Medicine, ATTIKON University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Mimma Rizzo
- Division of Medical Oncology, A.O.U. Consorziale Policlinico Di Bari, Bari, Italy
| | - Francesco Massari
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Roberto Iacovelli
- Medical Oncology Unit, Fondazione Policlinico A. Gemelli IRCCS, Rome, Italy
| | | |
Collapse
|
2
|
Xiong B, Liu W, Liu Y, Chen T, Lin A, Song J, Qu L, Luo P, Jiang A, Wang L. A Multi-Omics Prognostic Model Capturing Tumor Stemness and the Immune Microenvironment in Clear Cell Renal Cell Carcinoma. Biomedicines 2024; 12:2171. [PMID: 39457484 PMCID: PMC11504857 DOI: 10.3390/biomedicines12102171] [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: 08/06/2024] [Revised: 09/11/2024] [Accepted: 09/20/2024] [Indexed: 10/28/2024] Open
Abstract
Background: Cancer stem-like cells (CSCs), a distinct subset recognized for their stem cell-like abilities, are intimately linked to the resistance to radiotherapy, metastatic behaviors, and self-renewal capacities in tumors. Despite their relevance, the definitive traits and importance of CSCs in the realm of oncology are still not fully comprehended, particularly in the context of clear cell renal cell carcinoma (ccRCC). A comprehensive understanding of these CSCs' properties in relation to stemness, and their impact on the efficacy of treatment and resistance to medication, is of paramount importance. Methods: In a meticulous research effort, we have identified new molecular categories designated as CRCS1 and CRCS2 through the application of an unsupervised clustering algorithm. The analysis of these subtypes included a comprehensive examination of the tumor immune environment, patterns of metabolic activity, progression of the disease, and its response to immunotherapy. In addition, we have delved into understanding these subtypes' distinctive clinical presentations, the landscape of their genomic alterations, and the likelihood of their response to various pharmacological interventions. Proceeding from these insights, prognostic models were developed that could potentially forecast the outcomes for patients with ccRCC, as well as inform strategies for the surveillance of recurrence after treatment and the handling of drug-resistant scenarios. Results: Compared with CRCS1, CRCS2 patients had a lower clinical stage/grading and a better prognosis. The CRCS2 subtype was in a hypoxic state and was characterized by suppression and exclusion of immune function, which was sensitive to gefitinib, erlotinib, and saracatinib. The constructed prognostic risk model performed well in both training and validation cohorts, helping to identify patients who may benefit from specific treatments or who are at risk of recurrence and drug resistance. A novel therapeutic target, SAA2, regulating neutrophil and fibroblast infiltration, and, thus promoting ccRCC progression, was identified. Conclusions: Our findings highlight the key role of CSCs in shaping the ccRCC tumor microenvironment, crucial for therapy research and clinical guidance. Recognizing tumor stemness helps to predict treatment efficacy, recurrence, and drug resistance, informing treatment strategies and enhancing ccRCC patient outcomes.
Collapse
Affiliation(s)
- Beibei Xiong
- Department of Oncology, The First People’s Hospital of Shuangliu District, Chengdu 610200, China;
| | - Wenqiang Liu
- Department of Urology, Changhai Hospital, Navel Medical University (Second Military Medical University), Shanghai 200433, China; (W.L.); (Y.L.); (T.C.); (J.S.)
| | - Ying Liu
- Department of Urology, Changhai Hospital, Navel Medical University (Second Military Medical University), Shanghai 200433, China; (W.L.); (Y.L.); (T.C.); (J.S.)
| | - Tong Chen
- Department of Urology, Changhai Hospital, Navel Medical University (Second Military Medical University), Shanghai 200433, China; (W.L.); (Y.L.); (T.C.); (J.S.)
| | - Anqi Lin
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China; (A.L.); (P.L.)
| | - Jiaao Song
- Department of Urology, Changhai Hospital, Navel Medical University (Second Military Medical University), Shanghai 200433, China; (W.L.); (Y.L.); (T.C.); (J.S.)
| | - Le Qu
- Department of Urology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China;
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China; (A.L.); (P.L.)
| | - Aimin Jiang
- Department of Urology, Changhai Hospital, Navel Medical University (Second Military Medical University), Shanghai 200433, China; (W.L.); (Y.L.); (T.C.); (J.S.)
| | - Linhui Wang
- Department of Urology, Changhai Hospital, Navel Medical University (Second Military Medical University), Shanghai 200433, China; (W.L.); (Y.L.); (T.C.); (J.S.)
| |
Collapse
|
3
|
Rioja P, Rey-Cardenas M, De Velasco G. Targeting HIF-2α and anemia: A therapeutic breakthrough for clear-cell renal cell carcinoma. Cancer Treat Rev 2024; 129:102801. [PMID: 39032449 DOI: 10.1016/j.ctrv.2024.102801] [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: 05/19/2024] [Revised: 07/07/2024] [Accepted: 07/09/2024] [Indexed: 07/23/2024]
Abstract
Renal cell carcinoma (RCC) is a heterogenous disease which the incidence is increasing worldwide. The identification and understanding of the role of the Von Hipple Lindau (VHP) in regulating the hypoxia-inducible factor signaling pathway has revolutionized the treatment of this disease. Belzutifan is an oral hypoxia-inducible factor (HIF)-2α inhibitor, which has demonstrated efficacy in treating von Hippel-Lindau (VHL) disease and for the treatment of adults with RCC who experienced disease progression after PD-1/PD-L1- and VEGFR-targeted therapies. One of the most common adverse effect of this drug is anemia; however, it is treatment is not well known. This review summarizes role of the VHL-HIF pathway in ccRCC aroused the interest of targeting HIF activity, the history of belzutifan development and their relationship to anemia as well as propose a management algorithm.
Collapse
Affiliation(s)
- Patricia Rioja
- Department of Medical Oncology, National Institute of Neoplastic Diseases, Lima, Peru.
| | - M Rey-Cardenas
- Department of Medical Oncology, Gustave Roussy, Paris Saclay University, Villejuif, France
| | - Guillermo De Velasco
- Department of Medical Oncology, University Hospital 12 de Octubre, Instituto de investigación (imas12), Madrid, Spain
| |
Collapse
|
4
|
Omery BM, Rojas RF. FDG-Avid Metastatic Non-Clear Cell Renal Cell Carcinoma Presenting as Intussusception. Clin Nucl Med 2024; 49:864-865. [PMID: 38934462 DOI: 10.1097/rlu.0000000000005359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/28/2024]
Abstract
ABSTRACT A 47-year-old man with a history of metastatic non-clear cell left renal cell carcinoma, unclassified subtype, status post left radical nephrectomy 4 years prior, and treated with immunotherapy for approximately 2½ years, presented for 18 F-FDG PET/CT exam 7 months after immunotherapy was stopped. A contrast-enhanced CT exam performed 3 weeks prior demonstrated a new small bowel intussusception in the left upper quadrant. The PET/CT demonstrated focal FDG uptake in the segment of small bowel involved in the intussusception. Pathology from small bowel resection demonstrated metastatic renal cell carcinoma.
Collapse
Affiliation(s)
- Bilal M Omery
- From the Department of Radiology, Aurora St Luke's Medical Center, Milwaukee, WI
| | | |
Collapse
|
5
|
Cano Garcia C, Hoeh B, Mandal S, Banek S, Klümper N, Schmucker P, Hahn O, Mattigk A, Ellinger J, Cox A, Becker P, Zeuschner P, Zengerling F, Erdmann K, Buerk BT, Kalogirou C, Flegar L. First-Line Immune Combination Therapies for Nonclear Cell Versus Clear Cell Metastatic Renal Cell Carcinoma: Real-World Multicenter Data From Germany. Clin Genitourin Cancer 2024; 22:102112. [PMID: 38825563 DOI: 10.1016/j.clgc.2024.102112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 05/01/2024] [Accepted: 05/02/2024] [Indexed: 06/04/2024]
Abstract
INTRODUCTION The aim was to compare treatment outcomes of clear cell metastatic renal cell carcinoma (ccmRCC) versus non-ccmRCC (nccmRCC) patients who received first-line immune combination therapies. MATERIALS AND METHODS Within our retrospective multi-institutional consecutive database of eight tertiary-care centers, we identified mRCC patients treated with first-line immune combination therapies between 11/2017 and 12/2022. Using log-rank analysis and multivariable Cox regression, we tested for differences in overall survival (OS) and progression-free survival (PFS) of nccmRCC versus ccmRCC patients. Covariables consisted of age at diagnosis, sex, International Metastatic Renal Cell Carcinoma Database Consortium risk groups, Eastern Cooperative Oncology Group status, and sarcomatoid feature. RESULTS Of 289 study patients, 39 (13%) patients harbored nccmRCC. Median OS was 37 months versus not reached for ccmRCC versus nccmRCC patients (P = .6). Median PFS was 13 versus 15 months (P = .9). Multivariable Cox regression models did not identify nccmRCC as an independent predictor of higher overall mortality in mRCC patients (hazard ratio [HR]: 1.23; P = .6) or a higher progression rate (HR: 1.0; P = 1.0). CONCLUSION In our real-world multi-institutional study, no differences in OS and PFS between ccmRCC and nccmRCC patients receiving first-line immune combination treatment were observed, even after adjustment for important patient and tumor characteristics. More prospective trials in nccmRCC patients are needed.
Collapse
Affiliation(s)
- Cristina Cano Garcia
- Department of Urology, University Hospital Frankfurt, Goethe University Frankfurt am Main, Frankfurt, Germany.
| | - Benedikt Hoeh
- Department of Urology, University Hospital Frankfurt, Goethe University Frankfurt am Main, Frankfurt, Germany
| | - Subhajit Mandal
- Department of Urology, Philipps-University Marburg, Marburg, Germany
| | - Severine Banek
- Department of Urology, University Hospital Frankfurt, Goethe University Frankfurt am Main, Frankfurt, Germany
| | - Niklas Klümper
- Department of Urology, University Hospital Bonn (UKB), 53127 Bonn, Germany; Institute of Experimental Oncology, University Hospital Bonn (UKB), Bonn, Germany
| | - Philipp Schmucker
- Department of Urology and Paediatric Urology, Julius Maximilians University Medical Center of Würzburg, Würzburg, Germany
| | - Oliver Hahn
- Department of Urology and Paediatric Urology, Julius Maximilians University Medical Center of Würzburg, Würzburg, Germany
| | - Angelika Mattigk
- Department of Urology and Paediatric Urology, University Hospital Ulm, Ulm, Germany
| | - Jörg Ellinger
- Department of Urology, University Hospital Bonn (UKB), 53127 Bonn, Germany
| | - Alexander Cox
- Department of Urology, University Hospital Bonn (UKB), 53127 Bonn, Germany
| | - Philippe Becker
- Department of Urology and Paediatric Urology, Saarland University, Homburg/Saar, Germany
| | - Philip Zeuschner
- Department of Urology and Paediatric Urology, Saarland University, Homburg/Saar, Germany
| | | | - Kati Erdmann
- Department of Urology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Bjoern Thorben Buerk
- Department of Urology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Charis Kalogirou
- Department of Urology and Paediatric Urology, Julius Maximilians University Medical Center of Würzburg, Würzburg, Germany
| | - Luka Flegar
- Department of Urology, Philipps-University Marburg, Marburg, Germany
| |
Collapse
|
6
|
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
|
7
|
Piccinelli ML, Barletta F, Tappero S, Cano Garcia C, Incesu RB, Morra S, Scheipner L, Tian Z, Luzzago S, Mistretta FA, Ferro M, Saad F, Shariat SF, Ahyai S, Longo N, Tilki D, Chun FKH, Terrone C, Briganti A, de Cobelli O, Musi G, Karakiewicz PI. Development and External Validation of a Novel Nomogram Predicting Cancer-specific Mortality-free Survival in Surgically Treated Papillary Renal Cell Carcinoma Patients. Eur Urol Focus 2023; 9:799-806. [PMID: 37024421 DOI: 10.1016/j.euf.2023.03.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 03/08/2023] [Accepted: 03/23/2023] [Indexed: 04/07/2023]
Abstract
BACKGROUND Accurate prediction of cancer control outcomes in renal cell carcinoma (RCC) patients is important for counselling, follow-up planning, and selection of appropriate adjuvant trial designs. OBJECTIVE To develop and externally validate a novel contemporary population-based model for predicting cancer-specific mortality-free survival (CSM-FS) in surgically treated papillary RCC (papRCC) patients and to compare it with established risk categories (Leibovich 2018). DESIGN, SETTING, AND PARTICIPANTS Within the Surveillance, Epidemiology, and End Results database (2004-2019), we identified surgically treated papRCC patients (n = 3978). The population was randomly divided into development (50%, n = 1989) and external validation (50%, n = 1989) cohorts. Of the external validation cohort, 97% (n = 1930) of patients were included in a head-to-head comparison of the Leibovich 2018 risk categories addressing nonmetastatic patients. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS Univariable Cox regression models tested the statistical significance in the prediction of CSM-FS. The most parsimonious model with the best validation metrics was selected as the multivariable nomogram. Accuracy, calibration, and decision curve analyses (DCAs) tested the Cox regression-based nomogram, as well as the Leibovich 2018 risk categories in the external validation cohort. RESULTS AND LIMITATIONS Age at diagnosis, grade, T stage, N stage, and M stage qualified for inclusion in the novel nomogram. In external validation, the accuracy of the novel nomogram was 0.83 at 5 yr and 0.80 at 10 yr. In nonmetastatic patients, 5- and 10-yr accuracy of the novel nomogram was 0.77 and 0.76, respectively. Conversely, 5- and 10-yr accuracy of the Leibovich 2018 risk categories was 0.70 and 0.66, respectively. The novel nomogram exhibited smaller departures from ideal predictions in calibration plots and higher net benefit in DCAs, when it was compared with the Leibovich 2018 risk categories. Limitations include the retrospective nature of the study, absence of a central pathological review, and inclusion of only North American patients. CONCLUSIONS The novel nomogram may represent a valuable clinical aid, when papRCC CSM-FS predictions are required. PATIENT SUMMARY We developed an accurate tool to predict death due to papillary kidney cancer in a North American population.
Collapse
Affiliation(s)
- Mattia Luca Piccinelli
- Cancer Prognostics and Health Outcomes Unit, Division of Urology, University of Montréal Health Center, Montréal, Québec, Canada; Department of Urology, IEO European Institute of Oncology, IRCCS, Milan, Italy; Università degli Studi di Milano, Milan, Italy
| | - Francesco Barletta
- Cancer Prognostics and Health Outcomes Unit, Division of Urology, University of Montréal Health Center, Montréal, Québec, Canada; Division of Experimental Oncology/Unit of Urology, URI, Urological Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Stefano Tappero
- Cancer Prognostics and Health Outcomes Unit, Division of Urology, University of Montréal Health Center, Montréal, Québec, Canada; Department of Urology, IRCCS Policlinico San Martino, Genova, Italy; Department of Surgical and Diagnostic Integrated Sciences (DISC), University of Genova, Genova, Italy
| | - Cristina Cano Garcia
- Cancer Prognostics and Health Outcomes Unit, Division of Urology, University of Montréal Health Center, Montréal, Québec, Canada; Department of Urology, University Hospital Frankfurt, Goethe University Frankfurt am Main, Frankfurt am Main, Germany
| | - Reha-Baris Incesu
- Cancer Prognostics and Health Outcomes Unit, Division of Urology, University of Montréal Health Center, Montréal, Québec, Canada; Martini-Klinik Prostate Cancer Center, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Simone Morra
- Cancer Prognostics and Health Outcomes Unit, Division of Urology, University of Montréal Health Center, Montréal, Québec, Canada; Department of Neurosciences, Science of Reproduction and Odontostomatology, University of Naples Federico II, Naples, Italy
| | - Lukas Scheipner
- Cancer Prognostics and Health Outcomes Unit, Division of Urology, University of Montréal Health Center, Montréal, Québec, Canada; Department of Urology, Medical University of Graz, Graz, Austria
| | - Zhe Tian
- Cancer Prognostics and Health Outcomes Unit, Division of Urology, University of Montréal Health Center, Montréal, Québec, Canada
| | - Stefano Luzzago
- Department of Urology, IEO European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Haemato-Oncology, Università degli Studi di Milano, Milan, Italy
| | - Francesco A Mistretta
- Department of Urology, IEO European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Haemato-Oncology, Università degli Studi di Milano, Milan, Italy
| | - Matteo Ferro
- Department of Urology, IEO European Institute of Oncology, IRCCS, Milan, Italy
| | - Fred Saad
- Cancer Prognostics and Health Outcomes Unit, Division of Urology, University of Montréal Health Center, Montréal, Québec, Canada
| | - Shahrokh F Shariat
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Department of Urology, Weill Cornell Medical College, New York, NY, USA; Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX, USA; Hourani Center of Applied Scientific Research, Al-Ahliyya Amman University, Amman, Jordan
| | - Sascha Ahyai
- Department of Urology, Medical University of Graz, Graz, Austria
| | - Nicola Longo
- Department of Neurosciences, Science of Reproduction and Odontostomatology, University of Naples Federico II, Naples, Italy
| | - Derya Tilki
- Martini-Klinik Prostate Cancer Center, University Hospital Hamburg-Eppendorf, Hamburg, Germany; Department of Urology, University Hospital Hamburg-Eppendorf, Hamburg, Germany; Department of Urology, Koc University Hospital, Istanbul, Turkey
| | - Felix K H Chun
- Department of Urology, University Hospital Frankfurt, Goethe University Frankfurt am Main, Frankfurt am Main, Germany
| | - Carlo Terrone
- Department of Urology, IRCCS Policlinico San Martino, Genova, Italy; Department of Surgical and Diagnostic Integrated Sciences (DISC), University of Genova, Genova, Italy
| | - Alberto Briganti
- Division of Experimental Oncology/Unit of Urology, URI, Urological Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Ottavio de Cobelli
- Department of Urology, IEO European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Haemato-Oncology, Università degli Studi di Milano, Milan, Italy
| | - Gennaro Musi
- Department of Urology, IEO European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Haemato-Oncology, Università degli Studi di Milano, Milan, Italy
| | - Pierre I Karakiewicz
- Cancer Prognostics and Health Outcomes Unit, Division of Urology, University of Montréal Health Center, Montréal, Québec, Canada
| |
Collapse
|
8
|
Piccinelli ML, Tappero S, Cano Garcia C, Barletta F, Incesu RB, Morra S, Scheipner L, Tian Z, Luzzago S, Mistretta FA, Ferro M, Saad F, Shariat SF, Ahyai S, Longo N, Tilki D, Briganti A, Chun FK, Terrone C, de Cobelli O, Musi G, Karakiewicz PI. Assessment of the VENUSS and GRANT Models for Individual Prediction of Cancer-specific Survival in Surgically Treated Nonmetastatic Papillary Renal Cell Carcinoma. EUR UROL SUPPL 2023; 53:109-115. [PMID: 37441347 PMCID: PMC10334233 DOI: 10.1016/j.euros.2023.05.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/12/2023] [Indexed: 07/15/2023] Open
Abstract
Background Guidelines recommend VENUSS and GRANT models for the prediction of cancer control outcomes after nephrectomy for nonmetastatic papillary renal cell carcinoma (pRCC). Objective To test the ability of VENUSS and GRANT models to predict 5-yr cancer-specific survival in a North American population. Design setting and participants For this retrospective study, we identified 4184 patients with unilateral surgically treated nonmetastatic pRCC in the Surveillance, Epidemiology, and End Results database (2004-2019). Outcome measurements and statistical analysis The original VENUSS and GRANT risk categories were applied to predict 5-yr cancer-specific survival. A cross-validation method was used to test the accuracy and calibration of the models and to conduct decision curve analyses for the study cohort. Results and limitations The VENUSS and GRANT categories represented independent predictors of cancer-specific mortality. On cross-validation, the accuracy of the VENUSS and GRANT risk categories was 0.73 and 0.65, respectively. Both models showed good calibration and performed better than random predictions in decision curve analysis. Limitations include the retrospective nature of the study and the absence of a central pathological review. Conclusion VENUSS risk categories fulfilled prognostic model criteria for predicting cancer-specific survival 5 yr after surgery in North American patients with nonmetastatic pRCC as recommended by guidelines. Conversely, GRANT risk categories did not. Thus, VENUSS risk categories represent an important tool for counseling, follow-up planning, and patient selection for appropriate adjuvant trials in pRCC. Patient summary We tested the ability of two validated methods (VENUSS and GRANT) to predict death due to papillary kidney cancer in a North American population. The VENUSS risk categories showed good performance in predicting 5-year cancer-specific survival.
Collapse
Affiliation(s)
- Mattia L. Piccinelli
- Cancer Prognostics and Health Outcomes Unit, Division of Urology, University of Montréal Health Center, Montréal, Canada
- Department of Urology, IRCSS European Institute of Oncology, Milan, Italy
- Università degli Studi di Milano, Milan, Italy
| | - Stefano Tappero
- Cancer Prognostics and Health Outcomes Unit, Division of Urology, University of Montréal Health Center, Montréal, Canada
- Department of Urology, IRCCS Policlinico San Martino, Genova, Italy
- Department of Surgical and Diagnostic Integrated Sciences, University of Genova, Genova, Italy
| | - Cristina Cano Garcia
- Cancer Prognostics and Health Outcomes Unit, Division of Urology, University of Montréal Health Center, Montréal, Canada
- Department of Urology, University Hospital Frankfurt, Goethe University Frankfurt am Main, Frankfurt am Main, Germany
| | - Francesco Barletta
- Cancer Prognostics and Health Outcomes Unit, Division of Urology, University of Montréal Health Center, Montréal, Canada
- Division of Experimental Oncology/Unit of Urology, Urological Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Reha-Baris Incesu
- Cancer Prognostics and Health Outcomes Unit, Division of Urology, University of Montréal Health Center, Montréal, Canada
- Martini-Klinik Prostate Cancer Center, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Simone Morra
- Cancer Prognostics and Health Outcomes Unit, Division of Urology, University of Montréal Health Center, Montréal, Canada
- Department of Neurosciences, Science of Reproduction and Odontostomatology, University of Naples Federico II, Naples, Italy
| | - Lukas Scheipner
- Cancer Prognostics and Health Outcomes Unit, Division of Urology, University of Montréal Health Center, Montréal, Canada
- Department of Urology, Medical University of Graz, Graz, Austria
| | - Zhe Tian
- Cancer Prognostics and Health Outcomes Unit, Division of Urology, University of Montréal Health Center, Montréal, Canada
| | - Stefano Luzzago
- Department of Urology, IRCSS European Institute of Oncology, Milan, Italy
- Department of Oncology and Haemato-Oncology, Università degli Studi di Milano, Milan, Italy
| | - Francesco A. Mistretta
- Department of Urology, IRCSS European Institute of Oncology, Milan, Italy
- Department of Oncology and Haemato-Oncology, Università degli Studi di Milano, Milan, Italy
| | - Matteo Ferro
- Department of Urology, IRCSS European Institute of Oncology, Milan, Italy
| | - Fred Saad
- Cancer Prognostics and Health Outcomes Unit, Division of Urology, University of Montréal Health Center, Montréal, Canada
| | - Shahrokh F. Shariat
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
- Department of Urology, Weill Cornell Medical College, New York, NY, USA
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Hourani Center of Applied Scientific Research, Al-Ahliyya Amman University, Amman, Jordan
| | - Sascha Ahyai
- Department of Urology, Medical University of Graz, Graz, Austria
| | - Nicola Longo
- Department of Neurosciences, Science of Reproduction and Odontostomatology, University of Naples Federico II, Naples, Italy
| | - Derya Tilki
- Martini-Klinik Prostate Cancer Center, University Hospital Hamburg-Eppendorf, Hamburg, Germany
- Department of Urology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
- Department of Urology, Koc University Hospital, Istanbul, Turkey
| | - Alberto Briganti
- Division of Experimental Oncology/Unit of Urology, Urological Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Felix K.H. Chun
- Department of Urology, University Hospital Frankfurt, Goethe University Frankfurt am Main, Frankfurt am Main, Germany
| | - Carlo Terrone
- Department of Urology, IRCCS Policlinico San Martino, Genova, Italy
- Department of Surgical and Diagnostic Integrated Sciences, University of Genova, Genova, Italy
| | - Ottavio de Cobelli
- Department of Urology, IRCSS European Institute of Oncology, Milan, Italy
- Department of Oncology and Haemato-Oncology, Università degli Studi di Milano, Milan, Italy
| | - Gennaro Musi
- Department of Urology, IRCSS European Institute of Oncology, Milan, Italy
- Department of Oncology and Haemato-Oncology, Università degli Studi di Milano, Milan, Italy
| | - Pierre I. Karakiewicz
- Cancer Prognostics and Health Outcomes Unit, Division of Urology, University of Montréal Health Center, Montréal, Canada
| |
Collapse
|
9
|
Piccinelli ML, Panunzio A, Tappero S, Cano Garcia C, Barletta F, Incesu RB, Tian Z, Luzzago S, Mistretta FA, Ferro M, Saad F, Shariat SF, Tilki D, Briganti A, Chun FK, Terrone C, Antonelli A, DE Cobelli O, Musi G, Karakiewicz PI. Cancer-specific mortality free survival rates in non-metastatic non-clear cell renal carcinoma patients at intermediate/high risk of recurrence. Minerva Urol Nephrol 2023; 75:319-328. [PMID: 37221827 DOI: 10.23736/s2724-6051.23.05151-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
BACKGROUND To date, five trials testing the effect of adjuvant systemic therapy in surgically treated non-metastatic renal cell carcinoma included patients with non-clear cell histology. We tested the effect of papillary vs. chromophobe histological subtype, stage, and grade on 10-year cancer-specific survival, in patients eligible for ≥1 such trial. METHODS We identified patients meeting ASSURE, SORCE, EVEREST, PROSPER, or RAMPART trial inclusion criteria in the SEER (2000-2018) database. Kaplan-Meier analyses estimated 10-year survival rates and multivariable Cox regression models tested for the independent predictor status of histological subtype, stage, and grade. RESULTS We identified 5465 (68%) papillary and 2562 (32%) chromophobe renal cell carcinoma patients. Cancer-specific survival rates at 10 years were 77% in papillary vs. 90% in chromophobe. In multivariable Cox regression models applied to papillary patients, cancer-specific mortality independent predictor status was reached for T3G3-4 (HR 2.9), T4Gany (HR 3.4), TanyN1G1-2 (HR 3.1), and TanyN1G3-4 (HR 8.0, P<0.001), relative to T1/2Gany. In multivariable Cox regression models applied to chromophobe patients, mortality independent predictor status was reached for T3G3-4 (HR 3.6), T4Gany (HR 14.0), TanyN1G1-2 (HR 5.7), and TanyN1G3-4 (HR 15.0, P<0.001), relative to T1/2Gany. CONCLUSIONS In surgically treated non-metastatic intermediate/high-risk renal cell carcinoma patients, papillary histologic subtype exhibited worse cancer-specific survival than chromophobe histologic subtype. Although stage and grade represented independent predictors in both histological subtype groups, the magnitude of their effect was invariably worse in chromophobe than in papillary patients. In consequence, papillary and chromophobe patients should be considered separate entities instead of being combined under the non-clear cell designation.
Collapse
Affiliation(s)
- Mattia L Piccinelli
- Division of Urology, Unit of Cancer Prognostics and Health Outcomes, University of Montréal Health Center, Montréal, Canada -
- Department of Urology, IEO IRCCS European Institute of Oncology, Milan, Italy -
- University of Milan, Milan, Italy -
| | - Andrea Panunzio
- Division of Urology, Unit of Cancer Prognostics and Health Outcomes, University of Montréal Health Center, Montréal, Canada
- Department of Urology, Azienda Ospedaliera Universitaria Integrata di Verona, University of Verona, Verona, Italy
| | - Stefano Tappero
- Division of Urology, Unit of Cancer Prognostics and Health Outcomes, University of Montréal Health Center, Montréal, Canada
- Department of Urology, IRCCS San Martino Policlinic Hospital, Genoa, Italy
- Department of Surgical and Diagnostic Integrated Sciences (DISC), University of Genoa, Genoa, Italy
| | - Cristina Cano Garcia
- Division of Urology, Unit of Cancer Prognostics and Health Outcomes, University of Montréal Health Center, Montréal, Canada
- Department of Urology, University Hospital of Frankfurt, Goethe University Frankfurt am Main, Frankfurt am Main, Germany
| | - Francesco Barletta
- Division of Urology, Unit of Cancer Prognostics and Health Outcomes, University of Montréal Health Center, Montréal, Canada
- Division of Experimental Oncology, Unit of Urology, Urological Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Reha-Baris Incesu
- Division of Urology, Unit of Cancer Prognostics and Health Outcomes, University of Montréal Health Center, Montréal, Canada
- Martini-Klinik Prostate Cancer Center, University Hospital of Hamburg-Eppendorf, Hamburg, Germany
| | - Zhe Tian
- Division of Urology, Unit of Cancer Prognostics and Health Outcomes, University of Montréal Health Center, Montréal, Canada
| | - Stefano Luzzago
- Department of Urology, IEO IRCCS European Institute of Oncology, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Francesco A Mistretta
- Department of Urology, IEO IRCCS European Institute of Oncology, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Matteo Ferro
- Department of Urology, IEO IRCCS European Institute of Oncology, Milan, Italy
| | - Fred Saad
- Division of Urology, Unit of Cancer Prognostics and Health Outcomes, University of Montréal Health Center, Montréal, Canada
| | - Shahrokh F Shariat
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
- Department of Urology, Weill Cornell Medical College, New York, NY, USA
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Hourani Center of Applied Scientific Research, Al-Ahliyya Amman University, Amman, Jordan
| | - Derya Tilki
- Martini-Klinik Prostate Cancer Center, University Hospital of Hamburg-Eppendorf, Hamburg, Germany
- Department of Urology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
- Department of Urology, Koc University Hospital, Istanbul, Türkiye
| | - Alberto Briganti
- Division of Experimental Oncology, Unit of Urology, Urological Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Felix K Chun
- Department of Urology, University Hospital of Frankfurt, Goethe University Frankfurt am Main, Frankfurt am Main, Germany
| | - Carlo Terrone
- Department of Urology, IRCCS San Martino Policlinic Hospital, Genoa, Italy
- Department of Surgical and Diagnostic Integrated Sciences (DISC), University of Genoa, Genoa, Italy
| | - Alessandro Antonelli
- Department of Urology, Azienda Ospedaliera Universitaria Integrata di Verona, University of Verona, Verona, Italy
| | - Ottavio DE Cobelli
- Department of Urology, IEO IRCCS European Institute of Oncology, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Gennaro Musi
- Department of Urology, IEO IRCCS European Institute of Oncology, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Pierre I Karakiewicz
- Division of Urology, Unit of Cancer Prognostics and Health Outcomes, University of Montréal Health Center, Montréal, Canada
| |
Collapse
|
10
|
Piccinelli ML, Morra S, Tappero S, Cano Garcia C, Barletta F, Incesu RB, Scheipner L, Baudo A, Tian Z, Luzzago S, Mistretta FA, Ferro M, Saad F, Shariat SF, Carmignani L, Ahyai S, Tilki D, Briganti A, Chun FKH, Terrone C, Longo N, de Cobelli O, Musi G, Karakiewicz PI. Critical Appraisal of Leibovich 2018 and GRANT Models for Prediction of Cancer-Specific Survival in Non-Metastatic Chromophobe Renal Cell Carcinoma. Cancers (Basel) 2023; 15:cancers15072155. [PMID: 37046815 PMCID: PMC10093654 DOI: 10.3390/cancers15072155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 03/22/2023] [Accepted: 04/03/2023] [Indexed: 04/08/2023] Open
Abstract
Within the Surveillance, Epidemiology, and End Results database (2000–2019), we identified 5522 unilateral surgically treated non-metastatic chromophobe kidney cancer (chRCC) patients. This population was randomly divided into development vs. external validation cohorts. In the development cohort, the original Leibovich 2018 and GRANT categories were applied to predict 5- and 10-year cancer-specific survival (CSS). Subsequently, a novel multivariable nomogram was developed. Accuracy, calibration and decision curve analyses (DCA) tested the Cox regression-based nomogram as well as the Leibovich 2018 and GRANT risk categories in the external validation cohort. The accuracy of the Leibovich 2018 and GRANT models was 0.65 and 0.64 at ten years, respectively. The novel prognostic nomogram had an accuracy of 0.78 at ten years. All models exhibited good calibration. In DCA, Leibovich 2018 outperformed the novel nomogram within selected ranges of threshold probabilities at ten years. Conversely, the novel nomogram outperformed Leibovich 2018 for other values of threshold probabilities. In summary, Leibovich 2018 and GRANT risk categories exhibited borderline low accuracy in predicting CSS in North American non-metastatic chRCC patients. Conversely, the novel nomogram exhibited higher accuracy. However, in DCA, all examined models exhibited limitations within specific threshold probability intervals. In consequence, all three examined models provide individual predictions that might be suboptimal and be affected by limitations determined by the natural history of chRCC, where few deaths occur within ten years from surgery. Further investigations regarding established and novel predictors of CSS and relying on large sample sizes with longer follow-up are needed to better stratify CSS in chRCC.
Collapse
Affiliation(s)
- Mattia Luca Piccinelli
- Cancer Prognostics and Health Outcomes Unit, Division of Urology, University of Montréal Health Center, Montréal, QC H2X 0A9, Canada
- Department of Urology, IEO European Institute of Oncology, IRCCS, 20141 Milan, Italy
- Department of Oncology and Haemato-Oncology, Università degli Studi di Milano, 20122 Milan, Italy
| | - Simone Morra
- Cancer Prognostics and Health Outcomes Unit, Division of Urology, University of Montréal Health Center, Montréal, QC H2X 0A9, Canada
- Department of Neurosciences, Science of Reproduction and Odontostomatology, University of Naples Federico II, 80131 Naples, Italy
| | - Stefano Tappero
- Cancer Prognostics and Health Outcomes Unit, Division of Urology, University of Montréal Health Center, Montréal, QC H2X 0A9, Canada
- Department of Urology, IRCCS Policlinico San Martino, 16132 Genova, Italy
- Department of Surgical and Diagnostic Integrated Sciences (DISC), University of Genova, 16148 Genova, Italy
| | - Cristina Cano Garcia
- Cancer Prognostics and Health Outcomes Unit, Division of Urology, University of Montréal Health Center, Montréal, QC H2X 0A9, Canada
- Department of Urology, University Hospital Frankfurt, Goethe University Frankfurt am Main, 39120 Frankfurt am Main, Germany
| | - Francesco Barletta
- Cancer Prognostics and Health Outcomes Unit, Division of Urology, University of Montréal Health Center, Montréal, QC H2X 0A9, Canada
- Division of Experimental Oncology, Unit of Urology, URI Urological Research Institute, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Reha-Baris Incesu
- Cancer Prognostics and Health Outcomes Unit, Division of Urology, University of Montréal Health Center, Montréal, QC H2X 0A9, Canada
- Martini-Klinik Prostate Cancer Center, University Hospital Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Lukas Scheipner
- Cancer Prognostics and Health Outcomes Unit, Division of Urology, University of Montréal Health Center, Montréal, QC H2X 0A9, Canada
- Department of Urology, Medical University of Graz, 8036 Graz, Austria
| | - Andrea Baudo
- Cancer Prognostics and Health Outcomes Unit, Division of Urology, University of Montréal Health Center, Montréal, QC H2X 0A9, Canada
- Department of Urology, IRCCS Policlinico San Donato, 20097 Milan, Italy
| | - Zhe Tian
- Cancer Prognostics and Health Outcomes Unit, Division of Urology, University of Montréal Health Center, Montréal, QC H2X 0A9, Canada
| | - Stefano Luzzago
- Department of Urology, IEO European Institute of Oncology, IRCCS, 20141 Milan, Italy
- Department of Oncology and Haemato-Oncology, Università degli Studi di Milano, 20122 Milan, Italy
| | - Francesco Alessandro Mistretta
- Department of Urology, IEO European Institute of Oncology, IRCCS, 20141 Milan, Italy
- Department of Oncology and Haemato-Oncology, Università degli Studi di Milano, 20122 Milan, Italy
| | - Matteo Ferro
- Department of Urology, IEO European Institute of Oncology, IRCCS, 20141 Milan, Italy
| | - Fred Saad
- Cancer Prognostics and Health Outcomes Unit, Division of Urology, University of Montréal Health Center, Montréal, QC H2X 0A9, Canada
| | - Shahrokh F. Shariat
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria
- Department of Urology, Weill Cornell Medical College, New York, NY 10065, USA
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Hourani Center of Applied Scientific Research, Al-Ahliyya Amman University, Amman 19328, Jordan
| | - Luca Carmignani
- Department of Urology, IRCCS Policlinico San Donato, 20097 Milan, Italy
- Department of Urology, IRCCS Ospedale Galeazzi-Sant’Ambrogio, 20157 Milan, Italy
| | - Sascha Ahyai
- Department of Urology, Medical University of Graz, 8036 Graz, Austria
| | - Derya Tilki
- Martini-Klinik Prostate Cancer Center, University Hospital Hamburg-Eppendorf, 20246 Hamburg, Germany
- Department of Urology, University Hospital Hamburg-Eppendorf, 20246 Hamburg, Germany
- Department of Urology, Koc University Hospital, 34010 Istanbul, Turkey
| | - Alberto Briganti
- Division of Experimental Oncology, Unit of Urology, URI Urological Research Institute, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Felix K. H. Chun
- Department of Urology, University Hospital Frankfurt, Goethe University Frankfurt am Main, 39120 Frankfurt am Main, Germany
| | - Carlo Terrone
- Department of Urology, IRCCS Policlinico San Martino, 16132 Genova, Italy
- Department of Surgical and Diagnostic Integrated Sciences (DISC), University of Genova, 16148 Genova, Italy
| | - Nicola Longo
- Department of Neurosciences, Science of Reproduction and Odontostomatology, University of Naples Federico II, 80131 Naples, Italy
| | - Ottavio de Cobelli
- Department of Urology, IEO European Institute of Oncology, IRCCS, 20141 Milan, Italy
- Department of Oncology and Haemato-Oncology, Università degli Studi di Milano, 20122 Milan, Italy
| | - Gennaro Musi
- Department of Urology, IEO European Institute of Oncology, IRCCS, 20141 Milan, Italy
- Department of Oncology and Haemato-Oncology, Università degli Studi di Milano, 20122 Milan, Italy
| | - Pierre I. Karakiewicz
- Cancer Prognostics and Health Outcomes Unit, Division of Urology, University of Montréal Health Center, Montréal, QC H2X 0A9, Canada
| |
Collapse
|
11
|
Lobo J, Ohashi R, Amin MB, Berney DM, Compérat EM, Cree IA, Gill AJ, Hartmann A, Menon S, Netto GJ, Raspollini MR, Rubin MA, Tan PH, Tickoo SK, Tsuzuki T, Turajlic S, Zhou M, Srigley JR, Moch H. WHO 2022 landscape of papillary and chromophobe renal cell carcinoma. Histopathology 2022; 81:426-438. [PMID: 35596618 DOI: 10.1111/his.14700] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 05/04/2022] [Accepted: 05/18/2022] [Indexed: 12/01/2022]
Abstract
The 5th edition of the WHO Classification of Tumours of the Urinary and Male Genital Systems contains relevant revisions and introduces a group of molecularly defined renal tumour subtypes. Herein we present the World Health Organization (WHO) 2022 perspectives on papillary and chromophobe renal cell carcinoma with emphasis on their evolving classification, differential diagnosis, and emerging entities. The WHO 2022 classification eliminated the type 1/2 papillary renal cell carcinoma (pRCC) subcategorization, given the recognition of frequent mixed tumour phenotypes and the existence of entities with a different molecular background within the type 2 pRCC category. Additionally, emerging entities such as biphasic squamoid alveolar RCC, biphasic hyalinising psammomatous RCC, papillary renal neoplasm with reverse polarity, and Warthin-like pRCC are included as part of the pRCC spectrum, while additional morphological and molecular data are being gathered. In addition to oncocytomas and chromophobe renal cell carcinoma (chRCC), a category of 'other oncocytic tumours' with oncocytoma/chRCC-like features has been introduced, including emerging entities, most with TSC/mTOR pathway alterations (eosinophilic vacuolated tumour and so-called 'low-grade' oncocytic tumour), deserving additional research. Eosinophilic solid and cystic RCC was accepted as a new and independent tumour entity. Finally, a highly reproducible and clinically relevant universal grading system for chRCC is still missing and is another niche of ongoing investigation. This review discusses these developments and highlights emerging morphological and molecular data relevant for the classification of renal cell carcinoma.
Collapse
Affiliation(s)
- João Lobo
- Department of Pathology, Portuguese Oncology Institute of Porto (IPOP), Porto, Portugal
- Cancer Biology and Epigenetics Group, Research Center of IPO Porto (GEBC CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center (P.CCC), Porto, Portugal
- Department of Pathology and Molecular Immunology, ICBAS-School of Medicine and Biomedical Sciences, University of Porto (ICBAS-UP), Porto, Portugal
| | - Riuko Ohashi
- Histopathology Core Facility, Niigata University Faculty of Medicine, Niigata, Japan
- Division of Molecular and Diagnostic Pathology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Mahul B Amin
- Department of Pathology and Laboratory Medicine, University of Tennessee Health Sciences Center, Memphis, USA
- Department of Urology, University of Southern California, Los Angeles, CA, USA
| | - Daniel M Berney
- Centre for Molecular Oncology, Barts and the London School of Medicine and Dentistry, London, UK
| | - Eva M Compérat
- Department of Pathology, Hôpital Tenon, Sorbonne University, Paris, France
| | - Ian A Cree
- International Agency for Research on Cancer (IARC), World Health Organization, Lyon, France
| | - Anthony J Gill
- Sydney Medical School, The University of Sydney, Sydney, Australia
- Cancer Diagnosis and Pathology Group, Kolling Institute of Medical Research, St Leonards, New South Wales, Australia
- NSW Health Pathology, Department of Anatomical Pathology, Royal North Shore Hospital, NSW Health Pathology, St Leonards, New South Wales, Australia
| | - Arndt Hartmann
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Santosh Menon
- Department of Pathology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - George J Netto
- Department of Pathology, University of Alabama, Birmingham, AL, USA
| | - Maria R Raspollini
- Histopathology and Molecular Diagnostics, Careggi University Hospital, Florence, Italy
| | - Mark A Rubin
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY, USA
- Department for BioMedical Research, University of Bern, Bern, Switzerland
- Englander Institute for Precision Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Puay Hoon Tan
- Division of Pathology, Singapore General Hospital, Singapore
| | - Satish K Tickoo
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Toyonori Tsuzuki
- Department of Surgical Pathology, Aichi Medical University Hospital, Nagakute, Japan
| | - Samra Turajlic
- Renal and Skin Units, The Royal Marsden Hospital NHS Foundation Trust, London, UK
- The Francis Crick Institute, London, UK
| | - Ming Zhou
- Department of Pathology, Tufts Medical Center, Boston, Massachusetts, USA
| | - John R Srigley
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Holger Moch
- Department of Pathology and Molecular Pathology, University and University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
12
|
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
|
13
|
Erdem S, Capitanio U, Campi R, Mir MC, Roussel E, Pavan N, Kara O, Klatte T, Kriegmair MC, Degirmenci E, Aydin R, Minervini A, Serni S, Berni A, Rebez G, Ozcan F. External validation of the VENUSS prognostic model to predict recurrence after surgery in non-metastatic papillary renal cell carcinoma: A multi-institutional analysis. Urol Oncol 2022; 40:198.e9-198.e17. [DOI: 10.1016/j.urolonc.2022.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 12/26/2021] [Accepted: 01/11/2022] [Indexed: 10/19/2022]
|
14
|
Bigot C, Boudier P, Ladoire S, Barthélémy P. Advanced nccRCC: what therapeutic options in 2022? Bull Cancer 2022; 109:2S39-2S46. [DOI: 10.1016/s0007-4551(22)00237-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
15
|
Choueiri TK, Albiges L, Atkins MB, Bakouny Z, Bratslavsky G, Braun DA, Haas NB, Haanen JB, Hakimi AA, Jewett MA, Jonasch E, Kaelin WG, Kapur P, Labaki C, Lewis B, McDermott DF, Pal SK, Pels K, Poteat S, Powles T, Rathmell WK, Rini BI, Signoretti S, Tannir NM, Uzzo RG, Hammers HJ. From Basic Science to Clinical Translation in Kidney Cancer: A Report from the Second Kidney Cancer Research Summit. Clin Cancer Res 2022; 28:831-839. [PMID: 34965942 PMCID: PMC9223120 DOI: 10.1158/1078-0432.ccr-21-3238] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 11/07/2021] [Accepted: 12/21/2021] [Indexed: 11/16/2022]
Abstract
The second Kidney Cancer Research Summit was held virtually in October 2020. The meeting gathered worldwide experts in the field of kidney cancer, including basic, translational, and clinical scientists as well as patient advocates. Novel studies were presented, addressing areas of unmet need related to different topics. These include novel metabolic targets, promising immunotherapeutic regimens, predictive genomic and transcriptomic biomarkers, and variant histologies of renal cell carcinoma (RCC). With the development of pioneering technologies, and an unprecedented commitment to kidney cancer research, the field has tremendously evolved. This perspective aims to summarize the different sessions of the conference, outline major advances in the understanding of RCC and discuss current challenges faced by the field.
Collapse
Affiliation(s)
- Toni K. Choueiri
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Laurence Albiges
- Department of Medical Oncology, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Michael B. Atkins
- Georgetown Lombardi Comprehensive Cancer Center, Washington, DC, USA
| | - Ziad Bakouny
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Gennady Bratslavsky
- Department of Urology, State University of New York (SUNY) Upstate Medical University, Syracuse, New York
| | - David A. Braun
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Naomi B. Haas
- Division of Hematology and Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - John B.A.G. Haanen
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - A Ari Hakimi
- Department of Surgery, Urology Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Michael A.S. Jewett
- Division of Urology, Department of Surgery, Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Eric Jonasch
- Department of Genitourinary Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - William G. Kaelin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Payal Kapur
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Chris Labaki
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | - David F. McDermott
- Division of Medical Oncology, Beth Israel Deaconess Medical Center, Dana-Farber/Harvard Cancer Center, Boston, Massachusetts
| | - Sumanta K. Pal
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Kevin Pels
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | - Thomas Powles
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - W. Kimryn Rathmell
- Department of Medicine, Vanderbilt University Medical Center (VUMC), Nashville, TN, USA
| | - Brian I. Rini
- Department of Medicine, Vanderbilt University Medical Center (VUMC), Nashville, TN, USA
| | - Sabina Signoretti
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Nizar M. Tannir
- Department of Genitourinary Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Robert G. Uzzo
- Department of Surgical Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Hans J. Hammers
- Division of Hematology-Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
16
|
An J, Packiam VT, Chennamadhavuni A, Richards J, Jain J, Mott SL, Garje R. Patient Characteristics and Survival Outcomes of Non-Metastatic, Non-Clear Cell Renal Cell Carcinoma. Front Oncol 2022; 11:786307. [PMID: 35083144 PMCID: PMC8786111 DOI: 10.3389/fonc.2021.786307] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 12/09/2021] [Indexed: 11/17/2022] Open
Abstract
Background Non-clear cell renal cell carcinoma (ccRCC) includes histologically and molecularly distinct subtypes such as papillary, chromophobe, collecting duct, and sarcomatoid RCC, with an incidence ranging from 20% to 25%. Oncologic outcomes and the role of adjuvant systemic therapy [vascular endothelial growth factor inhibitor (VEGFi) or immunotherapy] for non-ccRCC are not well-described. Objective To assess the incidence and survival outcomes of non-ccRCC subtypes in comparison to ccRCC. Methods The National Cancer Database was utilized to identify patients with non-metastatic RCC (T1–T4, N0–N1) between 2004 and 2015. The non-ccRCC cohort was further stratified by histologic subtype: papillary, chromophobe, sarcomatoid, and collecting duct RCC. Multivariable Cox regression models were used to compare overall survival (OS). Results The 5-year OS for chromophobe, papillary, clear cell, collecting duct, and sarcomatoid RCC was 91%, 82%, 81%, 44%, and 40%, respectively. After adjusting for clinicopathologic and treatment characteristics, there was no significant difference in OS between papillary RCC and ccRCC (p = 0.17). Patients with collecting duct and sarcomatoid subtypes were at over two times increased risk of death compared to patients with clear cell (p < 0.01 and p < 0.01, respectively). Conversely, patients with chromophobe RCC were at 36% decreased risk of death compared to ccRCC (p < 0.01). Conclusions This hospital-based analysis confirms that collecting duct and sarcomatoid histologic subtypes are uncommon and associated with poor survival after surgery when compared to the other RCC subtypes. Further studies are needed to evaluate the role of neoadjuvant and adjuvant systemic therapies in these subtypes to improve oncologic outcomes.
Collapse
Affiliation(s)
- Josiah An
- Division of Hematology, Oncology, Blood & Marrow Transplantation, Department of Internal Medicine, University of Iowa Hospitals and Clinics, Iowa City, IA, United States
| | - Vignesh T Packiam
- Department of Urology, University of Iowa Hospitals and Clinics, Iowa City, IA, United States
| | - Adithya Chennamadhavuni
- Division of Hematology, Oncology, Blood & Marrow Transplantation, Department of Internal Medicine, University of Iowa Hospitals and Clinics, Iowa City, IA, United States
| | - Jordan Richards
- Department of Urology, University of Iowa Hospitals and Clinics, Iowa City, IA, United States
| | - Jayanshu Jain
- Department of Internal Medicine, University of Iowa Hospitals and Clinics, Iowa City, IA, United States
| | - Sarah L Mott
- Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, Iowa City, IA, United States
| | - Rohan Garje
- Division of Hematology, Oncology, Blood & Marrow Transplantation, Department of Internal Medicine, University of Iowa Hospitals and Clinics, Iowa City, IA, United States
| |
Collapse
|
17
|
Rasmussen IML, Soerensen AV, Møller AK, Persson GF, Palshof JA, Taarnhøj GA, Pappot H. How to individualise oncological treatment of patients with metastatic non-clear cell renal cell carcinoma by using gene sequencing and Patient-Reported Outcomes: INDIGO study protocol (Preprint). JMIR Res Protoc 2022; 11:e36632. [PMID: 36107483 PMCID: PMC9523525 DOI: 10.2196/36632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 05/19/2022] [Accepted: 06/13/2022] [Indexed: 11/13/2022] Open
Abstract
Background No phase 3 studies have yet been conducted for patients with non–clear cell (CC) renal cell carcinoma (RCC) exclusively due to the rare occurrence of the disease and the heterogenicity in tumor morphology. Consequently, there is no evidence of the optimal treatment, and new approaches are needed. One approach is individualizing treatment based on the gene sequencing of tumor tissue. Additionally, recent studies involving the patient-reported outcomes (PROs) of patients treated for metastatic cancer have shown significant benefits for quality of life, median overall survival, and overall survival. The use of gene sequencing and PROs can be of great importance to patients with rare cancer types, including patients with non-CC RCC, and should be investigated in clinical trials, especially for cases where evidence based on phase 3 studies is difficult to obtain. Objective We describe the INDIGO study, in which patients, based on gene analyses, will be allocated into 4 treatment arms containing 14 treatments and use electronic PROs. We aim to improve the treatment of patients with non-CC RCC. The end points in the study will be the overall response rate (complete and partial) in the total patient population, which will be based on the RECIST (Response Evaluation Criteria in Solid Tumors) version 1.1 criteria, and the time to treatment failure. Methods INDIGO is a prospective phase 2 trial, and 30 patients will be enrolled. The patients will receive systemic treatment based on genetic analyses of their tumor tissue. All patients will receive electronic questionnaires in a dedicated app—a questionnaire regarding symptoms and side effects and another regarding health-related quality of life. Depending on the treatment regimen, the patients will be seen by a medical doctor every third, fourth, or sixth week, and the effect of the systemic treatment will be evaluated every 6 weeks via a computed tomography scan. The study has been approved by the Danish Medicines Agency and the National Committee on Health Research Ethics (approval number: H-19041833), complies with good clinical practice guidelines, follows the General Data Protection Regulation, and is registered at the Capital Region of Denmark. Results Recruitment started in March 2020, and at the time of submitting this paper (June 2022), a total of 9 patients have been enrolled. Conclusions We aim to explore methods for improving the treatment outcomes of patients with non-CC RCC, and the INDIGO study will contribute further data on personalized medicine for rare types of RCC and provide new knowledge on the active use of electronic PROs. Trial Registration ClinicalTrials.gov NCT04644432, https://clinicaltrials.gov/ct2/show/NCT04644432 ; European Union Drug Regulating Authorities Clinical Trials Database 2019-001316-38, https://tinyurl.com/2p8mb4aw International Registered Report Identifier (IRRID) DERR1-10.2196/36632
Collapse
Affiliation(s)
- Ida Marie Lind Rasmussen
- Department of Oncology, Copenhagen University Hospital Herlev, Herlev, Denmark
- Department of Clinical Medicine, University of Copenhagen, København, Denmark
| | - Anne Vest Soerensen
- Department of Oncology, Copenhagen University Hospital Herlev, Herlev, Denmark
| | | | - Gitte Fredberg Persson
- Department of Oncology, Copenhagen University Hospital Herlev, Herlev, Denmark
- Department of Clinical Medicine, University of Copenhagen, København, Denmark
| | | | | | - Helle Pappot
- Department of Clinical Medicine, University of Copenhagen, København, Denmark
- Department of Oncology, Rigshospitalet, København, Denmark
| |
Collapse
|
18
|
Pal SK, McGregor B, Suárez C, Tsao CK, Kelly W, Vaishampayan U, Pagliaro L, Maughan BL, Loriot Y, Castellano D, Srinivas S, McKay RR, Dreicer R, Hutson T, Dubey S, Werneke S, Panneerselvam A, Curran D, Scheffold C, Choueiri TK, Agarwal N. Cabozantinib in Combination With Atezolizumab for Advanced Renal Cell Carcinoma: Results From the COSMIC-021 Study. J Clin Oncol 2021; 39:3725-3736. [PMID: 34491815 PMCID: PMC8601305 DOI: 10.1200/jco.21.00939] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Sumanta K Pal
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, CA
| | - Bradley McGregor
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Cristina Suárez
- Medical Oncology, Vall d'Hebron Institute of Oncology (VHIO), Hospital Universitari Vall d'Hebron, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Che-Kai Tsao
- Tisch Cancer Institute, Mount Sinai Hospital, New York, NY
| | - William Kelly
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA
| | - Ulka Vaishampayan
- Karmanos Cancer Center, Wayne State University, Detroit, MI.,Current affiliation: Division of Hematology/Oncology, University of Michigan, Ann Arbor, MI
| | | | | | - Yohann Loriot
- Department of Cancer Medicine, Gustave Roussy Institute, INSERM 981, University Paris-Saclay, Villejuif, France
| | - Daniel Castellano
- Department of Medical Oncology, Hospital 12 de Octubre, Madrid, Spain
| | - Sandy Srinivas
- Division of Medical Oncology, Stanford University Medical Center, Stanford, CA
| | - Rana R McKay
- University of California San Diego, San Diego, CA
| | | | - Thomas Hutson
- Charles A. Sammons Cancer Center at Baylor University Medical Center, Dallas, TX
| | | | | | | | | | | | - Toni K Choueiri
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Neeraj Agarwal
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| |
Collapse
|
19
|
Bergmann L, Weber S, Hartmann A, Ahrens M. Pathology and systemic therapy of non-clear cell renal cell carcinoma: an overview. Expert Rev Anticancer Ther 2021; 21:1273-1286. [PMID: 34291700 DOI: 10.1080/14737140.2021.1959319] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Introduction: Non-clear cell renal cell carcinoma (nccRCC) represents a highly heterogenous group of kidney cancer entities. As most clinical trials predominantly include patients with clear cell RCC (ccRCC), nccRCC treatment guidelines are mainly extrapolated from recommendations in ccRCC. Here, we review and elucidate current data on the pathologic classification and treatment of nccRCC.Areas covered: This article gives an overview of the WHO classification of RCC, showing the histological diversity of nccRCC and focusing particularly on entities first characterized since 2016, their specific molecular behavior and their role as indicators for hereditary cancer syndromes. In this context, we discuss the available data on nccRCC treatment oprtions such as tyrosine kinase inhibitors, mammalian target of rapamycin inhibitors, cytotoxic chemotherapy, and immune checkpoint inhibitors.Expert opinion: Although nccRCCs are relatively uncommon, entities of this type account for a subgroup of up to 20-25% of all RCCs. Advances in histopathology and molecular genetics, together with evidence gained from retrospective and prospective clinical data, have improved understanding of these tumors in recent years. Nevertheless, selective trials of current and novel therapies including new targeted agents in patients with nccRCC are urgently needed to further improve treatment guidelines.
Collapse
Affiliation(s)
- Lothar Bergmann
- Medical Clinic II, J.W. Goethe University, Frankfurt, Germany.,Private Praxis for Hematology/Oncology, Schifferstrasse, Frankfurt, Germany
| | - Sarah Weber
- Medical Clinic II, J.W. Goethe University, Frankfurt, Germany
| | - Arndt Hartmann
- Institute for Pathology, University Hospital, Erlangen, Nürnberg, Germany
| | - Marit Ahrens
- Medical Clinic II, J.W. Goethe University, Frankfurt, Germany
| |
Collapse
|
20
|
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
|
21
|
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
|
22
|
Zoumpourlis P, Genovese G, Tannir NM, Msaouel P. Systemic Therapies for the Management of Non-Clear Cell Renal Cell Carcinoma: What Works, What Doesn't, and What the Future Holds. Clin Genitourin Cancer 2021; 19:103-116. [PMID: 33358151 PMCID: PMC8169717 DOI: 10.1016/j.clgc.2020.11.005] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 10/31/2020] [Accepted: 11/18/2020] [Indexed: 02/06/2023]
Abstract
Non-clear cell renal cell carcinoma (nccRCC) is a broad term that refers to a diverse group of tumors, each with its own distinct biologic and therapeutic profile. The management of nccRCCs is often based on extrapolating data from clinical trials in the more common clear cell renal cell carcinoma, but our emerging prospective and retrospective clinical experience in nccRCC allows us to make more precise recommendations tailored to each histology. The systemic therapy options for metastatic nccRCC include targeted therapies such as tyrosine kinase inhibitors, immune checkpoint inhibitors, and, for specific rare subtypes, cytotoxic chemotherapy. Each nccRCC histology may respond differently to these regimens, which makes accurate pathologic diagnosis imperative. In the present review, we discuss the available clinical and biological data that can help guide systemic therapy recommendations for specific nccRCC subtypes.
Collapse
Affiliation(s)
| | - Giannicola Genovese
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX; Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Nizar M Tannir
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX.
| | - Pavlos Msaouel
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX; Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX; Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX.
| |
Collapse
|
23
|
Braun DA, Bakouny Z, Hirsch L, Flippot R, Van Allen EM, Wu CJ, Choueiri TK. Beyond conventional immune-checkpoint inhibition - novel immunotherapies for renal cell carcinoma. Nat Rev Clin Oncol 2021; 18:199-214. [PMID: 33437048 PMCID: PMC8317018 DOI: 10.1038/s41571-020-00455-z] [Citation(s) in RCA: 205] [Impact Index Per Article: 51.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/16/2020] [Indexed: 01/29/2023]
Abstract
The management of advanced-stage renal cell carcinoma (RCC) has been transformed by the development of immune-checkpoint inhibitors (ICIs). Nonetheless, most patients do not derive durable clinical benefit from these agents. Importantly, unlike other immunotherapy-responsive solid tumours, most RCCs have only a moderate mutational burden, and paradoxically, high levels of tumour CD8+ T cell infiltration are associated with a worse prognosis in patients with this disease. Building on the successes of antibodies targeting the PD-1 and CTLA4 immune checkpoints, multiple innovative immunotherapies are now in clinical development for the treatment of patients with RCC, including ICIs with novel targets, co-stimulatory pathway agonists, modified cytokines, metabolic pathway modulators, cell therapies and therapeutic vaccines. However, the successful development of such novel immune-based treatments and of immunotherapy-based combinations will require a disease-specific framework that incorporates a deep understanding of RCC immunobiology. In this Review, using the structure provided by the well-described cancer-immunity cycle, we outline the key steps required for a successful antitumour immune response in the context of RCC, and describe the development of promising new immunotherapies within the context of this framework. With this approach, we summarize and analyse the most encouraging targets of novel immune-based therapies within the RCC microenvironment, and review the landscape of emerging antigen-directed therapies for this disease.
Collapse
Affiliation(s)
- David A Braun
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ziad Bakouny
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Laure Hirsch
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Gustave Roussy, Villejuif, France
| | - Ronan Flippot
- Department of Medical Oncology, Gustave Roussy, Villejuif, France
| | - Eliezer M Van Allen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Catherine J Wu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Toni K Choueiri
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
24
|
Ishihara H, Tachibana H, Takagi T, Yoshida K, Kondo T, Tanabe K. Effect of improved systemic therapy on patient survival in metastatic non-clear-cell renal cell carcinoma. Int J Urol 2021; 28:605-607. [PMID: 33615577 DOI: 10.1111/iju.14523] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Hiroki Ishihara
- Department of Urology, Tokyo Women's Medical University, Tokyo, Japan
| | - Hidekazu Tachibana
- Department of Urology, Tokyo Women's Medical University Medical Center East, Tokyo, Japan
| | - Toshio Takagi
- Department of Urology, Tokyo Women's Medical University, Tokyo, Japan
| | - Kazuhiko Yoshida
- Department of Urology, Tokyo Women's Medical University, Tokyo, Japan
| | - Tsunenori Kondo
- Department of Urology, Tokyo Women's Medical University Medical Center East, Tokyo, Japan
| | - Kazunari Tanabe
- Department of Urology, Tokyo Women's Medical University, Tokyo, Japan
| |
Collapse
|
25
|
Mollica V, Franceschini T, Gruppioni E, Rizzo A, Ricci C, Schiavina R, Brunocilla E, Ardizzoni A, Fiorentino M, Giunchi F, Massari F. Broad spectrum mutational analysis of chromophobe renal cell carcinoma using next-generation sequencing. Pathol Res Pract 2021; 219:153350. [PMID: 33556910 DOI: 10.1016/j.prp.2021.153350] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 01/22/2021] [Accepted: 01/25/2021] [Indexed: 12/18/2022]
Abstract
BACKGROUND Chromophobe renal cell carcinoma (ChRCC) is a rare subtype of non-clear cell renal cell carcinoma. Due to its rarity, its molecular characterization as well as therapeutic targets are still not fully understood. METHODS We performed a next-generation sequencing analysis using the platform Ion PGM System on 20 retrospectively collected ChRCC cases with the aim of identify molecular biomarkers with potential prognostic value or that could have therapeutic implications. RESULTS We identified mutation onTP53, SMARCB1, RB1 and JAK3. The most frequently altered gene was TP53 (6/20, 30 % of cases). SMARCB1 mutation was found in 3 (15 %) patients and in all cases the mutational variant was p.T72 K, with known pathogenenic meaning. One (5%) patient presented a pathogenetic mutation of RB1. JAK3 was mutated in 1 (5%) patient and this mutation resulted to have uncertain pathogenetic significance. CONCLUSION ChRCC is a rare disease still not fully molecularly characterized. Next-generation sequencing analysis could be useful to identify potential mutation with prognostic value or that could be potential therapeutic targets.
Collapse
Affiliation(s)
- Veronica Mollica
- Division of Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni - 15, Bologna, Italy
| | - Tania Franceschini
- Department of Pathology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni - 15, Bologna, Italy
| | - Elisa Gruppioni
- Department of Pathology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni - 15, Bologna, Italy
| | - Alessandro Rizzo
- Division of Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni - 15, Bologna, Italy
| | - Costantino Ricci
- Department of Specialistic Diagnostic and Experimental Medicine, University of Bologna, Via Massarenti 9, 40138, Bologna, Italy
| | - Riccardo Schiavina
- Department of Urology, University of Bologna, S-Orsola-Malpighi Hospital, Italy
| | - Eugenio Brunocilla
- Department of Urology, University of Bologna, S-Orsola-Malpighi Hospital, Italy
| | - Andrea Ardizzoni
- Division of Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni - 15, Bologna, Italy
| | - Michelangelo Fiorentino
- Department of Specialistic Diagnostic and Experimental Medicine, University of Bologna, Via Massarenti 9, 40138, Bologna, Italy.
| | - Francesca Giunchi
- Department of Pathology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni - 15, Bologna, Italy
| | - Francesco Massari
- Division of Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni - 15, Bologna, Italy.
| |
Collapse
|
26
|
Dudani S, de Velasco G, Wells JC, Gan CL, Donskov F, Porta C, Fraccon A, Pasini F, Lee JL, Hansen A, Bjarnason GA, Beuselinck B, Pal SK, Yuasa T, Kroeger N, Kanesvaran R, Reaume MN, Canil C, Choueiri TK, Heng DYC. Evaluation of Clear Cell, Papillary, and Chromophobe Renal Cell Carcinoma Metastasis Sites and Association With Survival. JAMA Netw Open 2021; 4:e2021869. [PMID: 33475752 PMCID: PMC7821027 DOI: 10.1001/jamanetworkopen.2020.21869] [Citation(s) in RCA: 142] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
IMPORTANCE There exists considerable biological and clinical variability between histologic variants of metastatic renal cell carcinoma (mRCC). Data reporting on patterns of metastasis in histologic variants of mRCC are sparse. OBJECTIVE To characterize sites of metastasis and their association with survival across the 3 most common histologic variants of mRCC: clear cell (ccRCC), papillary (pRCC), and chromophobe (chrRCC). DESIGN, SETTING, AND PARTICIPANTS In this multicenter, international cohort study, the International mRCC Database Consortium (IMDC) database was used to identify consecutive patients starting systemic therapy for mRCC between 2002 and 2019. Patients with mixed histologic subtype were excluded. Statistical analysis was performed from February to June 2020. EXPOSURES Data regarding histologic subtype and sites of metastatic involvement at the time of first systemic therapy initiation were collected. MAIN OUTCOMES AND MEASURES The primary outcomes were prevalence of metastatic site involvement and overall survival (OS) from time of systemic therapy initiation. Patients with multiple sites of metastatic involvement were included in analyses of all groups to which they had metastases. RESULTS A total of 10 105 patients were eligible for analysis. Median (interquartile range) age at diagnosis was 60 (53-67) years, 7310 (72.4%) were men and 8526 (84.5%) underwent nephrectomy. Of these, 9252 (92%) had ccRCC, 667 (7%) had pRCC, and 186 (2%) had chrRCC. The median number of sites of metastasis was 2 (range, 0-7). In ccRCC, the most common sites of metastasis were lung (70%; 6189 of 8804 patients [448 missing]), lymph nodes (45%; 3874 of 8655 patients [597 missing]), bone (32%; 2847 of 8817 patients [435 missing]), liver (18%; 1560 of 8804 [448 missing]), and adrenal gland (10%; 678 of 6673 patients [2579 missing]). Sites of metastasis varied between subtypes. Lung, adrenal, brain, and pancreatic metastases were more frequent in ccRCC, lymph node involvement was more common in pRCC, and liver metastases were more frequent in chrRCC. Median OS for ccRCC varied by site of metastatic involvement, ranging between 16 months (95% CI, 13.7-18.8 months) for the pleura and 50 months (95% CI, 41.1-55.5 months) for the pancreas. Compared with ccRCC, patients with pRCC tended to have lower OS, regardless of metastatic site. CONCLUSIONS AND RELEVANCE Sites of metastatic involvement differ according to histologic subtype in mRCC and are associated with OS. These data highlight the clinical and biological variability between histologic subtypes of mRCC. Patterns of metastatic spread may reflect differences in underlying disease biology. Further work to investigate differences in immune, molecular, and genetic profiles between metastatic sites and histologic subtypes is encouraged.
Collapse
Affiliation(s)
- Shaan Dudani
- Tom Baker Cancer Centre, University of Calgary, Calgary, Alberta, Canada
| | - Guillermo de Velasco
- Department of Medical Oncology, University Hospital 12 de Octubre, Madrid, Spain
| | - J Connor Wells
- Tom Baker Cancer Centre, University of Calgary, Calgary, Alberta, Canada
| | - Chun Loo Gan
- Tom Baker Cancer Centre, University of Calgary, Calgary, Alberta, Canada
| | - Frede Donskov
- Department of Oncology,Aarhus University Hospital, Aarhus, Denmark
| | - Camillo Porta
- Department of Internal Medicine, University of Pavia, Pavia, Italy
- Now with Department of Oncology, University of Bari Aldo Moro, Bari, Italy
| | | | - Felice Pasini
- Oncologia Medica Ospedale Santa Maria della Misericordia, Rovigo, Italy
| | - Jae Lyun Lee
- Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Aaron Hansen
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Georg A Bjarnason
- Division of Medical Oncology, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Benoit Beuselinck
- Leuven Cancer Institute, University Hospitals Leuven, Leuven, Belgium
| | - Sumanta K Pal
- City of Hope Comprehensive Cancer Center, Duarte, California
| | - Takeshi Yuasa
- Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Nils Kroeger
- Department of Urology, University Medicine Greifswald, Greifswald, Germany
| | | | - M Neil Reaume
- The Ottawa Hospital Cancer Centre, University of Ottawa, Ottawa, Ontario, Canada
| | - Christina Canil
- The Ottawa Hospital Cancer Centre, University of Ottawa, Ottawa, Ontario, Canada
| | - Toni K Choueiri
- Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Daniel Y C Heng
- Tom Baker Cancer Centre, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
27
|
Park JJ, Kellezi O, Hamasha R, Ali A, Alva AS. Immunotherapy in metastatic sarcomatoid renal cell carcinoma: A single institution experience. Cancer Treat Res Commun 2020; 25:100251. [PMID: 33310369 DOI: 10.1016/j.ctarc.2020.100251] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 10/27/2020] [Accepted: 10/31/2020] [Indexed: 01/20/2023]
Abstract
INTRODUCTION Immune checkpoint inhibitors (CPIs) were recently approved in advanced clear cell renal cell carcinoma (RCC) and could be a promising option for metastatic RCC with sarcomatoid differentiation (sRCC) which otherwise carry a poor prognosis. We sought to compare outcomes between patients who received immunotherapy (IO) including CPIs or high dose interleukin-2 (HD IL2) for metastatic sRCC versus those who did not. PATIENTS AND METHODS We performed a single-center retrospective data analysis of 44 consecutive sRCC patients with any percentage of sarcomatoid differentiation from our institutional RCC database of whom 34 received IO and 10 patients did not. RESULTS Baseline variables between the two groups were not significantly different except for a greater percentage of patients with ≥40% sarcomatoid differentiation in the non-IO cohort. At a median follow-up of 27.6 months, patients treated with IO had a median overall survival of 57.6 months compared to 6.6 months in patients not treated with IO (p = 0.0002). Overall response rates (ORR) between the IO and non-IO group were 35.3% and 0% respectively (p = 0.06). When IO was given in the 1st line setting, the ORR was 25.0%, as compared to 44.4% in the 2nd line setting and beyond though limitations of small sample sizes apply. Immune-related adverse events (IRAE) occurred in 38.2% of patients in the IO group, with grade 3 events (mostly gastrointestinal) in 20.6% with no grade 4 or 5 events. IRAEs led to interruption or discontinuation of immunotherapy in 26.5%. CONCLUSION Our results support IO as an effective therapeutic option for patients with metastatic sarcomatoid RCC. Further study of various IO regimens, including those affecting the interleukin-2 signaling pathway, and their efficacy in neoadjuvant and adjuvant settings are warranted in sRCC.
Collapse
Affiliation(s)
- Joseph J Park
- Department of Internal Medicine, University of Michigan, 7310 Rogel Comprehensive Cancer Center, Ann Arbor, MI, 48109, USA
| | - Olesia Kellezi
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Reema Hamasha
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Alicia Ali
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Ajjai S Alva
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
28
|
Dai C, Huang J, Li Y, Zhang S, Tan Q, Hou J, Tang Q, Hu X, Guo J, Zeng M, Wang H, Zhou J. Tumor contour irregularity on preoperative imaging: a practical and useful prognostic parameter for papillary renal cell carcinoma. Eur Radiol 2020; 31:3745-3753. [PMID: 33211144 DOI: 10.1007/s00330-020-07456-7] [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: 07/26/2020] [Revised: 09/07/2020] [Accepted: 11/03/2020] [Indexed: 11/25/2022]
Abstract
OBJECTIVES To illustrate tumor contour irregularity on preoperative imaging with a practical method and further determine its value in predicting disease-free survival (DFS) in patients with pRCC (papillary renal cell carcinoma). METHODS We performed a retrospective single-institution review of 267 Chinese pRCC patients between March 2009 and May 2019. Contour irregularity on cross-section was classified into smooth but distorted margin, unsmooth and sharply nodular margin, and blurred margin. Then, the ratio of the cross-section numbers of irregularity and the total tumor was defined as the contour irregular degree (CID). Cox regression and Kaplan-Meier analysis were performed to analyze the impact of CID on DFS. Then, the prognostic performance of CID was compared with pRCC risk stratification published by Leibovich et al. RESULTS: The median follow-up was 45 months (IQR: 23-69), in which 27 (10%) patients had metastasis or recurrence. Observed DFS rates were 95%, 90%, and 88% at 1, 3, and 5 years. The CID was an independent prognostic factor of DFS (HR = 1.048, 95% CI = 1.029-1.068, p < 0.001). The Kaplan-Meier plot showed that high-risk patients (CID ≥ 50%) tended to have a significantly shorter DFS (p < 0.001). The CID and Leibovich's pRCC model for DFS prediction had a C-index of 0.934 (95% CI = 0.907-0.961) and 0.833 (95% CI = 0.739-0.927) respectively. CONCLUSIONS With our standard and practical method, the CID can be a reliable imaging marker for DFS prediction in patients with pRCC. KEY POINTS • The updated contour irregularity was an independent parameter for predicting disease-free survival in patients with pRCC. • High-risk pRCC patients (contour irregular degree ≥ 50%) tended to have a shorter disease-free survival. • Tumor contour irregularity in pRCC risk stratification outperformed Leibovich's model from our cohort.
Collapse
Affiliation(s)
- Chenchen Dai
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai Medical Imaging Institute, No180, Fenglin Road, Xuhui District, Shanghai, China
| | - Jiaqi Huang
- Department of Urology, Zhongshan Hospital, Fudan University, No180, Fengling Road, Xuhui District, Shanghai, China
| | - Yaohui Li
- Department of Urology, Zhongshan Hospital, Fudan University, No180, Fengling Road, Xuhui District, Shanghai, China
| | - Sihong Zhang
- Department of Urology, Zhongshan Hospital, Fudan University, No180, Fengling Road, Xuhui District, Shanghai, China
| | - Qinxuan Tan
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai Medical Imaging Institute, No180, Fenglin Road, Xuhui District, Shanghai, China
| | - Jun Hou
- Department of Pathology, Zhongshan Hospital, Fudan University, No180, Fenglin Road, Xuhui District, Shanghai, China
| | - Qiying Tang
- Department of Radiology, Xiamen Branch, Zhongshan Hospital, Fudan University, No 668, Jinhu Road, Huli District, Xiamen, Fujian province, China
| | - Xiaoyi Hu
- Department of Urology, Zhongshan Hospital, Fudan University, No180, Fengling Road, Xuhui District, Shanghai, China
| | - Jianming Guo
- Department of Urology, Zhongshan Hospital, Fudan University, No180, Fengling Road, Xuhui District, Shanghai, China
| | - Mengsu Zeng
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai Medical Imaging Institute, No180, Fenglin Road, Xuhui District, Shanghai, China
| | - Hang Wang
- Department of Urology, Zhongshan Hospital, Fudan University, No180, Fengling Road, Xuhui District, Shanghai, China.
| | - Jianjun Zhou
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai Medical Imaging Institute, No180, Fenglin Road, Xuhui District, Shanghai, China. .,Department of Radiology, Xiamen Branch, Zhongshan Hospital, Fudan University, No 668, Jinhu Road, Huli District, Xiamen, Fujian province, China.
| |
Collapse
|
29
|
Choueiri TK, Heng DYC, Lee JL, Cancel M, Verheijen RB, Mellemgaard A, Ottesen LH, Frigault MM, L’Hernault A, Szijgyarto Z, Signoretti S, Albiges L. Efficacy of Savolitinib vs Sunitinib in Patients With MET-Driven Papillary Renal Cell Carcinoma: The SAVOIR Phase 3 Randomized Clinical Trial. JAMA Oncol 2020; 6:1247-1255. [PMID: 32469384 PMCID: PMC7260692 DOI: 10.1001/jamaoncol.2020.2218] [Citation(s) in RCA: 120] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 05/02/2020] [Indexed: 01/16/2023]
Abstract
Importance Papillary renal cell carcinoma (PRCC) is the most common type of non-clear cell RCC. Because some cases of PRCC are MET-driven, MET inhibition could be a targeted treatment approach. In previous studies, savolitinib (AZD6094, HMPL-504, volitinib), a highly selective MET-tyrosine kinase inhibitor, demonstrated antitumor activity in this patient group. Objective To determine whether savolitinib is a better treatment option for this patient population, vs standard of care, sunitinib. Design, Setting, and Participants The SAVOIR phase 3, open-label, randomized clinical trial was a multicenter study carried out in 32 centers in 7 countries between July 2017 and the data cutoff in August 2019. Overall, 360 to 450 patients were to be screened to randomize approximately 180 patients. Patients were adults with MET-driven (centrally confirmed), metastatic PRCC, with 1 or more measurable lesions. Exclusion criteria included prior receipt of sunitinib or MET inhibitor treatment. Overall, 254 patients were screened. Interventions Patients received 600 mg of savolitinib orally once daily (qd), or 50 mg of sunitinib orally qd for 4 weeks, followed by 2 weeks without treatment. Main Outcomes and Measures The primary end point was progression-free survival (PFS, assessed by investigator and confirmed by blinded independent central review). Secondary end points included overall survival (OS), objective response rate (ORR), duration of response, and safety/tolerability. Results At data cutoff, 60 patients were randomized (savolitinib n = 33; sunitinib n = 27); most patients had chromosome 7 gain (savolitinib, 30 [91%]; sunitinib, 26 [96%]) and no prior therapy (savolitinib, 28 [85%]; sunitinib, 25 [93%]). For savolitinib and sunitinib, 4 (12%) and 10 (37%) patients were women, and the median (range) age was 60 (23-78) and 65 (31-77) years, respectively. Following availability of external data on PFS with sunitinib in patients with MET-driven disease, study enrollment was closed. Progression-free survival, OS, and ORR were numerically greater with savolitinib vs sunitinib. Median PFS was not statistically different between the 2 groups: 7.0 months (95% CI, 2.8-not calculated) for savolitinib and 5.6 months (95% CI, 4.1-6.9) for sunitinib (hazard ratio [HR], 0.71; 95% CI, 0.37-1.36; P = .31). For savolitinib and sunitinib respectively, grade 3 or higher adverse events (AEs) were reported in 14 (42%) and 22 (81%) of patients and AE-related dose modifications in 10 (30%) and 20 (74%). After discontinuation, 12 (36%) and 5 (19%) of patients on savolitinib and sunitinib respectively, received subsequent anticancer therapy. Conclusions and Relevance Although patient numbers and follow-up were limited, savolitinib demonstrated encouraging efficacy vs sunitinib, with fewer grade 3 or higher AEs and dose modifications. Further investigation of savolitinib as a treatment option for MET-driven PRCC is warranted. Trial Registration ClinicalTrials.gov Identifier: NCT03091192.
Collapse
Affiliation(s)
- Toni K. Choueiri
- Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Daniel Y. C. Heng
- Department of Medical Oncology, Tom Baker Cancer Center, University of Calgary, Calgary, Canada
| | - Jae Lyun Lee
- Asan Medical Center and University of Ulsan College of Medicine, Seoul, South Korea
| | | | | | | | | | | | | | | | - Sabina Signoretti
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Laurence Albiges
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Department of Cancer Medicine, Institut Gustave Roussy, Villejuif, France
| |
Collapse
|
30
|
Iacovelli R, Ciccarese C, Bria E, Bracarda S, Porta C, Procopio G, Tortora G. Patients with sarcomatoid renal cell carcinoma - re-defining the first-line of treatment: A meta-analysis of randomised clinical trials with immune checkpoint inhibitors. Eur J Cancer 2020; 136:195-203. [PMID: 32712550 DOI: 10.1016/j.ejca.2020.06.008] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 05/29/2020] [Accepted: 06/09/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND Sarcomatoid renal cell carcinoma (sRCC) represents a rare form of renal cell carcinoma marked by an aggressive biology, poor prognosis and little benefit from anti-angiogenic targeted therapy. More promising results come from the recent therapeutic strategy based on immune checkpoint inhibitor (ICI) combinations. MATERIALS AND METHODS For this meta-analysis, we searched MEDLINE/PubMed, the Cochrane Library and American Society of Medical Oncology (ASCO) Meeting abstracts for phase II or III randomised clinical trials. Data extraction was conducted according to the Preferred Reporting Items for Systematic Review and Meta-Analysis statement. The hazard ratios (HRs) for progression-free survival (PFS) and overall survival (OS) with the relative 95% confidence intervals were extracted from studies. Summary HRs were calculated using random- or fixed-effects models, depending on the heterogeneity of the included studies. RESULTS Four studies were selected for final analysis, including 467 patients (226 treated in with ICI combinations and 241 received sunitinib in the control arms). ICI-based combinations were associated with an improved PFS and OS compared with sunitinib, with a reduction of more than 40% of progression (HR = 0.56; p < 0.0001) and mortality (HR = 0.56; p = 0.001) risk. Moreover, ICI-based combinations are associated with a objective response rate (ORR) of more than 50% (versus 20% with sunitinib), corresponding to a doubled risk of achieving an ORR compared with controls (relative risk [RR] = 2.15; p < 0.00001). Finally, immunotherapy significantly increased the possibility to obtain complete responses (RR = 8.15, p = 0.0002) with an incidence of 11%. CONCLUSION Our data support the efficacy of ICI-based combinations for sRCC therapy, redefining the first-line treatment.
Collapse
Affiliation(s)
- Roberto Iacovelli
- Medical Oncology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy.
| | - Chiara Ciccarese
- Medical Oncology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy.
| | - Emilio Bria
- Medical Oncology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy.
| | - Sergio Bracarda
- Department of Oncology, Azienda Ospedaliera Santa Maria, Terni, 05100 Terni, Italy.
| | - Camillo Porta
- Department of Biomedical Sciences and Human Oncology, University of Bari "A. Moro", 70121 Bari, Italy.
| | - Giuseppe Procopio
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale Dei Tumori di Milano, Milan, Italy.
| | - Giampaolo Tortora
- Medical Oncology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy
| |
Collapse
|
31
|
de Vries-Brilland M, Gross-Goupil M, Seegers V, Boughalem E, Beuselinck B, Thibault C, Chevreau C, Ladoire S, Barthélémy P, Negrier S, Borchiellini D, Huillard O, Geoffrois L, Gravis G, Saldana C, Thiery-Vuillemin A, Escudier B, Ravaud A, Albiges L. Are immune checkpoint inhibitors a valid option for papillary renal cell carcinoma? A multicentre retrospective study. Eur J Cancer 2020; 136:76-83. [PMID: 32653774 DOI: 10.1016/j.ejca.2020.02.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 02/07/2020] [Indexed: 10/23/2022]
Abstract
BACKGROUND Papillary renal cell carcinoma (pRCC) is the most common non-clear cell RCC (nccRCC). Pivotal studies evaluating immune checkpoint inhibitors mostly excluded nccRCC. The aim of this retrospective and multicentre study was to evaluate the activity of programmed death-1 (PD-1)/ programmed death-ligand 1 (PD-L1) inhibitors specifically in metastatic pRCC. METHODS The primary end-point was time to treatment failure (TTF). Secondary endpoints included objective response rate (ORR), overall survival (OS) and treatment-related adverse events (TRAEs). RESULTS From 02/2016 to 01/2019, 57 patients with pRCC were included. Histology included 16 (28%) type 1 pRCC, 34 (60%) type 2 pRCC and 7 (12%) unclassified pRCC. Treatment with immune checkpoint inhibitors was used in the first-line setting in 4 patients (7%), in the second-line setting in 32 patients (56%) and in the third-line setting or more in 21 patients (37%). With a median follow-up of 12 months (95% confidence interval [CI]: 9.9-21.0), the median TTF was 3.1 months (95% CI: 2.7-5.0). Among the 55 patients evaluable for ORR, best response was complete response/partial response in 6 patients (11%), stable disease in 18 patients (33%) and progressive disease in 31 patients (56%). The median OS was 14.6 months (95% CI: 9.0- not reached). TRAEs of grade III-IV were noted in 6 patients (10%) leading to treatment discontinuation, and no grade V TRAEs were observed. CONCLUSION PD-1/PD-L1 inhibitors exhibit limited activity as monotherapy in this pRCC population, which remains an unmet need. Our findings underline the need for further prospective clinical trials evaluating immune checkpoint inhibitor combinations in patients with pRCC.
Collapse
Affiliation(s)
- Manon de Vries-Brilland
- Department of Medical Oncology, Integrated Centers of Oncology (ICO) Paul Papin, Angers, France
| | - Marine Gross-Goupil
- Department of Medical Oncology, Hôpital Saint-André, University of Bordeaux-CHU, Bordeaux, France
| | - Valérie Seegers
- Biostatistics Department, Integrated Centers of Oncology (ICO) Paul Papin, Angers, France
| | - Elouen Boughalem
- Department of Medical Oncology, Integrated Centers of Oncology (ICO) Paul Papin, Angers, France
| | - Benoit Beuselinck
- University Hospitals Leuven, Leuven Cancer Institute, Leuven, Belgium
| | | | | | - Sylvain Ladoire
- Department of Medical Oncology, Center GF Leclerc, Dijon, France
| | | | | | | | - Olivier Huillard
- Cochin Hospital, Paris Descartes University, AP-HP, CARPEM, Immunomodulatory Therapies Multidisciplinary Study Group, Paris, France
| | - Lionnel Geoffrois
- Department of Medical Oncology, Institut de Cancérologie de Lorraine, Vandœuvre-Lès-Nancy, France
| | | | - Carolina Saldana
- Oncology Department, Hôpital Henri Mondor, APHP, Créteil, France
| | | | - Bernard Escudier
- Medical Oncology, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Alain Ravaud
- Department of Medical Oncology, Hôpital Saint-André, University of Bordeaux-CHU, Bordeaux, France
| | - Laurence Albiges
- Medical Oncology, Gustave Roussy, Université Paris-Saclay, Villejuif, France.
| |
Collapse
|
32
|
Mollica V, Rizzo A, Massari F. Re: Toni K. Choueiri, Daniel Y.C. Heng, Jae Lyun Lee, et al. Efficacy of Savolitinib vs Sunitinib in Patients With MET-Driven Papillary Renal Cell Carcinoma: The SAVOIR Phase 3 Randomized Clinical Trial. JAMA Oncol. In press. https://doi.org/10.1001/jamaoncol.2020.2218: SAVOIR: From Own Goal to Winning Goal? Eur Urol Oncol 2020; 3:561-562. [PMID: 32653414 DOI: 10.1016/j.euo.2020.06.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Accepted: 06/22/2020] [Indexed: 01/31/2023]
Affiliation(s)
- Veronica Mollica
- Division of Oncology, S.Orsola-Malpighi Hospital, Bologna, Italy
| | - Alessandro Rizzo
- Division of Oncology, S.Orsola-Malpighi Hospital, Bologna, Italy
| | | |
Collapse
|
33
|
Bergmann L, Grünwald V, Maute L, Grimm MO, Weikert S, Schleicher J, Klotz T, Greiner J, Flörcken A, Hartmann A, Gauler T. A Randomized Phase IIa Trial with Temsirolimus versus Sunitinib in Advanced Non-Clear Cell Renal Cell Carcinoma: An Intergroup Study of the CESAR Central European Society for Anticancer Drug Research-EWIV and the Interdisciplinary Working Group on Renal Cell Cancer (IAGN) of the German Cancer Society. Oncol Res Treat 2020; 43:333-339. [PMID: 32541143 DOI: 10.1159/000508450] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 05/06/2020] [Indexed: 11/19/2022]
Abstract
BACKGROUND Non-clear cell renal cell cancers (nccRCC) are rare entities, and the optimal therapy in metastatic disease has still to be defined. METHODS In this small prospectively randomized phase IIa multicenter trial, we investigated temsirolimus (TEM) versus sunitinib (SUN) as first-line therapy in patients with metastatic nccRCC. The patients were randomized 1:1 to either TEM in a dose of 25 mg i.v. once a week or SUN with 50 mg p.o. daily for 4 weeks on and 2 weeks off. Primary endpoint was progression-free survival (PFS). In total, 22 patients were included with predominantly papillary RCC (16/22) followed by chromophobe RCC and others. RESULTS The male to female ratio was 16:6. The tumor control rate (CR + PR + SD) was 58% for TEM and 90% for SUN-treated patients. There was also a trend for improved PFS with 9.3 versus 13.2 months (HR 1.64; 95% CI 0.65-4.18) in favor of SUN. There was no trend for overall survival. CONCLUSIONS Despite this trial had to be terminated earlier due to low recruitment, the results match the other studies published so far with the mTOR inhibitor everolimus and SUN, which show a trend in favor of SUN for ORR and PFS.
Collapse
Affiliation(s)
- Lothar Bergmann
- Medical Clinic II, J.W. Goethe University, Frankfurt, Germany,
| | - Viktor Grünwald
- Hematology and Medical Oncology, Medizinische Hochschule Hannover, Hannover, Germany
| | - Luise Maute
- Medical Clinic II, J.W. Goethe University, Frankfurt, Germany
| | | | - Steffen Weikert
- Clinic for Urology, Vivantes Humboldt-Klinikum, Berlin, Germany
| | - Jan Schleicher
- Clinic for Hematology and Oncology, Klinikum Stuttgart, Stuttgart, Germany
| | - Theodor Klotz
- Clinic for Urology and Andrology, Kliniken Nordoberpfalz AG, Weiden, Germany
| | - Jochen Greiner
- Clinic for Hematology and Oncology, Diakonie Klinikum, Stuttgart, Germany
| | - Anne Flörcken
- Department of Hematology, Oncology, and Tumor Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Campus Virchow-Klinikum, Berlin, Germany.,Clinic for Hematology and Oncology, Charité, Berlin, Germany
| | - Arndt Hartmann
- Institut for Pathology, University Hospital, Erlangen-Nürnberg, Germany
| | - Thomas Gauler
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen of the University Duisburg-Essen, Essen, Germany
| |
Collapse
|
34
|
Abstract
Variant histology renal cell carcinoma (vRCC) encompasses rare non-clear cell subtypes that have long been associated with poor prognosis and minimal response to therapies targeting vascular endothelial growth factor and its receptor. Molecular advances have helped classify vRCC into distinct entities and identify putative targetable driver alterations, such as MET in papillary subtypes. More have since been identified in other vRCC subtypes, including alterations of tumor metabolism, chromatin remodeling genes, cell-cycle genes, and inactivation of tumor suppressors such as TP53 or NF2. New targeted therapies, as well as immune checkpoint inhibitors, have been in development and yielded encouraging results. Collaborative clinical trials will be an essential step toward better implementation of these regimens in clinical practice.
Collapse
|
35
|
Les cancers du rein non à cellules claires : caractéristiques clinico-biologiques et prise en charge thérapeutique hors chirurgie. Bull Cancer 2020; 107:S56-S65. [DOI: 10.1016/s0007-4551(20)30279-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
36
|
Safety and efficacy of CDX-014, an antibody-drug conjugate directed against T cell immunoglobulin mucin-1 in advanced renal cell carcinoma. Invest New Drugs 2020; 38:1807-1814. [PMID: 32472319 DOI: 10.1007/s10637-020-00945-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 05/06/2020] [Indexed: 01/08/2023]
Abstract
CDX-014 is an antibody-drug conjugate directed against TIM-1, a surface marker highly expressed in renal cell carcinoma (RCC) and ovarian carcinoma. This phase I, first-in-human trial was conducted to evaluate the safety and preliminary activity of CDX-014 in patients with advanced refractory RCC, following a dose-escalation and dose expansion design. CDX-014 was administered intravenously at doses ranging from 0.15 to 2.0 mg/kg every 2 or 3 weeks until progression or unacceptable toxicity. Sixteen patients received at least one dose of CDX-014. The maximum tolerated dose was not identified. Most frequent adverse grade 1 or 2 adverse events included nausea (38%), fatigue, alopecia, elevation of AST and decreased appetite (25% each). Adverse events of grade 3 or more included hyperglycemia (19%), urosepsis (6%), and one multi-organ failure (6%) responsible for one treatment-related death. Two patients discontinued therapy for adverse events including fatigue grade 2 and urosepsis grade 4. CDX-014 showed antitumor activity with one prolonged partial response and a clinical benefit rate (objective response or stable disease >6 months) of 31%. The two patients that exhibited the most marked tumor shrinkage had high TIM-1 expression on tumor tissue. Overall, CDX-014 exhibited a manageable toxicity profile and early signs of activity, supporting further evaluation of antibody-drug conjugates in patients with advanced RCC and potentially other TIM-1 expressing cancers. Trial registration https://clinicaltrials.gov/ct2/show/NCT02837991 NCT02837991; July 20, 2016.
Collapse
|
37
|
Latest progress in molecular biology and treatment in genitourinary tumours. Clin Transl Oncol 2020; 22:2175-2195. [PMID: 32440915 DOI: 10.1007/s12094-020-02373-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 05/01/2020] [Indexed: 12/24/2022]
Abstract
The management of genitourinary cancer, including bladder, prostate, renal and testicular cancer, has evolved dramatically in recent years due to a better understanding of tumour genetic mutations, alterations in molecular pathways, and to the development of new kinds of drugs such as targeted therapies and immunotherapies. In the field of immunotherapy, new drugs focused on stimulating, enhancing and modulating the immune system to detect and destroy cancer, have been recently discovered. Research in oncology moves quickly and new data of great relevance for clinical practice are communicated every year. For this reason, a group of experts, focused exclusively on the treatment of genitourinary tumours and who get together every year in the BestGU conference to assess the latest progress in this field have summarized the most important advances in a single review, along with a critical assessment of whether these results should alter daily clinical practice.
Collapse
|
38
|
Liu YJ, Houldsworth J, Emmadi R, Dyer L, Wolff DJ. Assessing Genomic Copy Number Alterations as Best Practice for Renal Cell Neoplasia: An Evidence-Based Review from the Cancer Genomics Consortium Workgroup. Cancer Genet 2020; 244:40-54. [PMID: 32434132 DOI: 10.1016/j.cancergen.2020.04.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 04/06/2020] [Accepted: 04/07/2020] [Indexed: 02/05/2023]
Abstract
Renal cell neoplasia are heterogeneous with diverse histology, genetic alterations, and clinical behavior that are diagnosed mostly on morphologic features. The Renal Cell Neoplasia Workgroup of the Cancer Genomics Consortium systematically evaluated peer-reviewed literature on genomic studies of renal cell carcinoma (RCC), including clear cell RCC, papillary RCC, chromophobe RCC, and the translocation RCC involving TFE3, TFEB and MITF rearrangements, as well as benign oncocytoma, which together comprise about 95% of all renal cell neoplasia. The Workgroup curated recurrent copy number alterations (CNAs), copy-neutral loss-of-heterozygosity (cnLOH), rearrangements, and mutations, found in each subtype and assigned clinical relevance according to established criteria. In clear cell RCC, loss of 3p has a disease-initiating role and most likely also in progression with mutations detected in VHL and other genes mapped to this arm, and loss of 9p and/or 14q has well-substantiated prognostic utility. Gain of chromosomes 7 and 17 are hallmark CNAs of papillary RCC, but patterns of other CNAs as detected by chromosomal microarray analysis (CMA) afford sub-classification into Type 1 and 2 with prognostic value, and for further sub-stratification of Type 2. Inherent chromosome loss in chromophobe RCC as detected by CMA is useful for distinguishing the eosinophilic variant from benign oncocytoma which in contrast exhibits few CNAs or rearranged CCND1, but share mitochondrial DNA mutations. In morphologically atypical RCCs, rearrangement of TFE3 and TFEB should be considered in the differential diagnosis, portending an aggressive RCC subtype. Overall, this evidence-based review provides a validated role for assessment of CNAs in renal cell neoplasia in the clinical setting to assist in renal cell neoplasm diagnosis and sub-classification within subtypes that is integral to the management of patients, from small incidentally found renal masses to larger surgically resected specimens, and simultaneously identify the presence of key alterations portending outcome in malignant RCC subtypes.
Collapse
Affiliation(s)
- Yajuan J Liu
- Departments of Pathology and Laboratory Medicine, University of Washington School of Medicine, 1959 NE Pacific Street, Seattle, WA 98195.
| | - Jane Houldsworth
- Department of Pathology, Molecular and Cell Based Medicine, Icahn School of Medicine at Mount Sinai, The Mount Sinai Health System, 1 Gustave Levy Place, New York, NY 10029.
| | - Rajyasree Emmadi
- Department of Pathology, University of Illinois at Chicago, 840 S. Wood Street, Chicago, IL 60612
| | - Lisa Dyer
- Department of Pediatrics, Division of Human Genetics, University of Cincinnati, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, MLC 4006, Cincinnati, OH 45229-3039
| | - Daynna J Wolff
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, 165 Ashley Avenue, MSC 908, Charleston, SC 29425
| |
Collapse
|
39
|
Debien V, Thouvenin J, Lindner V, Barthélémy P, Lang H, Flippot R, Malouf GG. Sarcomatoid Dedifferentiation in Renal Cell Carcinoma: From Novel Molecular Insights to New Clinical Opportunities. Cancers (Basel) 2019; 12:E99. [PMID: 31906050 PMCID: PMC7016737 DOI: 10.3390/cancers12010099] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 12/19/2019] [Accepted: 12/20/2019] [Indexed: 01/10/2023] Open
Abstract
Sarcomatoid features in renal cell carcinoma (RCC) have long been associated with dismal prognosis and poor response to therapy, while biological mechanisms underpinning sarcomatoid dedifferentiation remained obscure. Several efforts have been conducted to break down the molecular profile of sarcomatoid RCC and investigate different targeted therapeutic approaches. Mutations enriched for in sarcomatoid RCC involve, notably, TP53, BAP1, cell cycle, and chromatin-remodeling genes. The immunological landscape of these tumors is also gradually being uncovered, showing frequent expression of programmed cell death ligand-1 (PD-L1) and high levels of tumor-infiltrating lymphocytes. These features may be major determinants for the activity of immune checkpoint inhibitors in this population, which has been confirmed by retrospective studies and subgroup analyses of large randomized phase 3 trials. Combinations based on PD-1/PD-L1 inhibition have demonstrated response rates and complete responses in >50% and >10% of patients in the first-line metastatic setting, respectively, with median overall survival exceeding two years. This remarkable improvement in outcomes effectively establishes immune checkpoint inhibitor combinations as a new standard of care in patients with sarcomatoid RCC. New research fields, including epigenetic regulations and tumor-microenvironment interactions, may further sharpen understanding of sarcomatoid RCC and advance therapeutic developments.
Collapse
Affiliation(s)
- Véronique Debien
- Department of Oncology, Institut de Cancérologie de Strasbourg, Hôpitaux Universitaires de Strasbourg, Université de Strasbourg, 67200 Strasbourg, France; (V.D.); (J.T.)
- Department of Cancer and Functional Genomics, Institute of Genetics and Molecular and Cellular Biology, CNRS/INSERM/UNISTRA, 67400 Illkirch, France;
| | - Jonathan Thouvenin
- Department of Oncology, Institut de Cancérologie de Strasbourg, Hôpitaux Universitaires de Strasbourg, Université de Strasbourg, 67200 Strasbourg, France; (V.D.); (J.T.)
- Department of Cancer and Functional Genomics, Institute of Genetics and Molecular and Cellular Biology, CNRS/INSERM/UNISTRA, 67400 Illkirch, France;
| | - Véronique Lindner
- Department of Pathology, Centre Hospitalier Universitaire Régional de Strasbourg, 67200 Strasbourg, France;
| | - Philippe Barthélémy
- Department of Cancer and Functional Genomics, Institute of Genetics and Molecular and Cellular Biology, CNRS/INSERM/UNISTRA, 67400 Illkirch, France;
| | - Hervé Lang
- Department of Urology, Centre Hospitalier Universitaire Régional de Strasbourg, 67000 Strasbourg, France;
| | - Ronan Flippot
- Department of Cancer Medicine, Gustave Roussy, 94800 Villejuif, France;
| | - Gabriel G. Malouf
- Department of Oncology, Institut de Cancérologie de Strasbourg, Hôpitaux Universitaires de Strasbourg, Université de Strasbourg, 67200 Strasbourg, France; (V.D.); (J.T.)
- Department of Cancer and Functional Genomics, Institute of Genetics and Molecular and Cellular Biology, CNRS/INSERM/UNISTRA, 67400 Illkirch, France;
| |
Collapse
|
40
|
Jeruc J, Čugura T, Tomažič A, Boštjančič E. MDM2-positive papillary sarcomatoid renal cell carcinoma: a potential diagnostic pitfall. Virchows Arch 2019; 476:783-786. [PMID: 31732813 DOI: 10.1007/s00428-019-02703-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 09/12/2019] [Accepted: 10/17/2019] [Indexed: 11/28/2022]
Abstract
Sarcomatoid renal cell carcinoma is a highly aggressive form of carcinoma, histologically showing both carcinomatous and mesenchymal component in different proportions. We present a case of advanced type 1 papillary sarcomatoid renal cell carcinoma infiltrating adjacent organs and showing positivity for MDM2 by immunohistochemistry and MDM2 amplification by fluorescence in situ hybridization. This finding, together with sarcomatoid morphology, poses a potential pitfall for diagnosis with dedifferentiated liposarcoma. MDM2 is known to be altered in various human sarcomas. Only recently, MDM2 alterations have been reported in carcinomas. The presented case illustrates the need of thorough sampling with clinic-pathological correlation before making a final diagnosis in sarcomatoid retroperitoneal tumours. Additionally, the potential clinical implications of MDM2 amplification in renal cell carcinoma are discussed.
Collapse
Affiliation(s)
- Jera Jeruc
- Institute of Pathology, Faculty of Medicine, University of Ljubljana, Korytkova 2, 1000, Ljubljana, Slovenia.
| | - Tanja Čugura
- Institute of Pathology, Faculty of Medicine, University of Ljubljana, Korytkova 2, 1000, Ljubljana, Slovenia
| | - Aleš Tomažič
- Department of Abdominal Surgery, University Medical Center Ljubljana, Zaloška cesta 7, 1000, Ljubljana, Slovenia
| | - Emanuela Boštjančič
- Institute of Pathology, Faculty of Medicine, University of Ljubljana, Korytkova 2, 1000, Ljubljana, Slovenia
| |
Collapse
|
41
|
Di Nunno V, Massari F, Mollica V, Cimadamore A, Santoni M, Cheng L, Lopez-Beltran A, Scarpelli M, Montironi R. Another one in the chamber: cabozantinib for patients with metastatic non clear cell renal cell carcinoma. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:S137. [PMID: 31576344 DOI: 10.21037/atm.2019.06.06] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
| | | | - Veronica Mollica
- Division of Oncology, S. Orsola-Malpighi Hospital, Bologna, Italy
| | - Alessia Cimadamore
- Section of Pathological Anatomy, Polytechnic University of the Marche Region, School of Medicine, United Hospitals, Ancona, Italy
| | | | - Liang Cheng
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | - Marina Scarpelli
- Section of Pathological Anatomy, Polytechnic University of the Marche Region, School of Medicine, United Hospitals, Ancona, Italy
| | - Rodolfo Montironi
- Section of Pathological Anatomy, Polytechnic University of the Marche Region, School of Medicine, United Hospitals, Ancona, Italy
| |
Collapse
|
42
|
McKay RR, Bossé D, Choueiri TK. Evolving Systemic Treatment Landscape for Patients With Advanced Renal Cell Carcinoma. J Clin Oncol 2018; 36:JCO2018790253. [PMID: 30372392 DOI: 10.1200/jco.2018.79.0253] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE To outline current practices and challenges in the systemic management of patients with advanced renal cell carcinoma (RCC). DESIGN We conducted a focused review of hallmark randomized controlled trials informing the systemic treatment of patients with RCC. We concentrated on trials informing the use of combination therapies, therapy in both treatment-naïve and previously treated patients, sequential treatment strategies, and schedules. RESULTS The systemic treatment of advanced RCC has experienced tremendous progress over the past 15 years. An improved understanding of the canonical pathways implicated in RCC pathogenesis has resulted in the development of molecularly targeted and immunotherapy options for patients. These therapies have replaced cytokine-based treatments as the standard of care for patients with advanced RCC. Until recently, sequential vascular endothelial growth factor (VEGF)-targeted therapy or VEGF-targeted therapy followed by mammalian target of rapamycin inhibition has been the prevailing treatment paradigm for patients. However, newer agents such as cabozantinib and nivolumab have challenged this traditional approach. In addition, combination treatments including nivolumab plus ipilimumab and atezolizumab plus bevacizumab have transformed the RCC treatment landscape, and other doublet combinations in clinical testing will likely continue to alter the treatment paradigm in RCC. Currently, factors that inform treatment selection between different therapy options include performance status, comorbidities, prognostic risk stratification, treatment adverse event profile, and mode of administration, with no Level I evidence for predictive biomarker use in clinic. CONCLUSIONS The treatment options for advanced RCC are rapidly evolving since the introduction of VEGF-targeted therapy, immunotherapy with checkpoint blockade and, more recently, combination regimens. Despite the success of these regimens, advanced RCC remains a largely incurable disease, and additional strategies are warranted.
Collapse
Affiliation(s)
- Rana R McKay
- Rana R. McKay, Moores Cancer Center, University of California San Diego, San Diego, CA; Dominick Bossé and Toni K. Choueiri, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA; Dominick Bossé, The Ottawa Hospital Cancer Center, University of Ottawa, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Dominick Bossé
- Rana R. McKay, Moores Cancer Center, University of California San Diego, San Diego, CA; Dominick Bossé and Toni K. Choueiri, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA; Dominick Bossé, The Ottawa Hospital Cancer Center, University of Ottawa, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Toni K Choueiri
- Rana R. McKay, Moores Cancer Center, University of California San Diego, San Diego, CA; Dominick Bossé and Toni K. Choueiri, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA; Dominick Bossé, The Ottawa Hospital Cancer Center, University of Ottawa, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| |
Collapse
|
43
|
McDermott DF, Carducci M. Progress in Kidney Cancer Outcomes Through Collaboration, Innovation, and Discovery. J Clin Oncol 2018; 36:JCO1801198. [PMID: 30372393 PMCID: PMC6299339 DOI: 10.1200/jco.18.01198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/06/2023] Open
Affiliation(s)
- David F. McDermott
- David F. McDermott, Dana-Farber/Harvard Cancer Center, Boston, MA; and Michael Carducci, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD
| | - Michael Carducci
- David F. McDermott, Dana-Farber/Harvard Cancer Center, Boston, MA; and Michael Carducci, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD
| |
Collapse
|