338 results on '"Martin H. Voss"'
Search Results
102. Telaglenastat Plus Cabozantinib or Everolimus for Advanced or Metastatic Renal Cell Carcinoma: An Open-Label Phase I Trial
- Author
-
Funda Meric-Bernstam, Nizar M. Tannir, Othon Iliopoulos, Richard J. Lee, Melinda L. Telli, Alice C. Fan, Angela DeMichele, Naomi B. Haas, Manish R. Patel, James J. Harding, Martin H. Voss, Taofeek K. Owonikoko, Bradley Carthon, Ramaprasad Srinivasan, Johanna C. Bendell, Yonchu Jenkins, Sam H. Whiting, Keith Orford, Mark K. Bennett, and Todd M. Bauer
- Subjects
Diarrhea ,Male ,Cancer Research ,Pyridines ,Kidney Neoplasms ,Article ,Oncology ,Antineoplastic Combined Chemotherapy Protocols ,Humans ,Anilides ,Female ,Everolimus ,Enzyme Inhibitors ,Carcinoma, Renal Cell ,Transaminases - Abstract
Purpose:Dual inhibition of glucose and glutamine metabolism results in synergistic anticancer effects in solid tumor models. Telaglenastat, an investigational, small-molecule, glutaminase inhibitor, exhibits modest single-agent activity in renal cell carcinoma (RCC) patients. This phase Ib trial evaluated telaglenastat plus cabozantinib or everolimus, agents known to impair glucose metabolism in patients with metastatic RCC (mRCC).Patients and Methods:mRCC patients received escalating doses of telaglenastat [400–800 mg per os (p.o.) twice daily] in a 3 + 3 design, plus either everolimus (10 mg daily p.o.; TelaE) or cabozantinib (60 mg daily p.o.; TelaC). Tumor response (RECISTv1.1) was assessed every 8 weeks. Endpoints included safety (primary) and antitumor activity.Results:Twenty-seven patients received TelaE, 13 received TelaC, with median 2 and 3 prior therapies, respectively. Treatment-related adverse events were mostly grades 1 to 2, most common including decreased appetite, anemia, elevated transaminases, and diarrhea with TelaE, and diarrhea, decreased appetite, elevated transaminases, and fatigue with TelaC. One dose-limiting toxicity occurred per cohort: grade 3 pruritic rash with TelaE and thrombocytopenia with TelaC. No maximum tolerated dose (MTD) was reached for either combination, leading to a recommended phase II dose of 800-mg telaglenastat twice daily with standard doses of E or C. TelaE disease control rate (DCR; response rate + stable disease) was 95.2% [20/21, including 1 partial response (PR)] among 21 patients with clear cell histology and 66.7% (2/3) for papillary. TelaC DCR was 100% (12/12) for both histologies [5/10 PRs as best response (3 confirmed) in clear cell].Conclusions:TelaE and TelaC showed encouraging clinical activity and tolerability in heavily pretreated mRCC patients.
- Published
- 2021
103. Axitinib plus Immuncheckpoint-Inhibitor: evidenz- und expertenbasierte Konsensusempfehlungen für die Behandlungsoptimierung und das Management von therapieassoziierten Nebenwirkungen
- Author
-
Rachel H. Giles, Viktor Grünwald, Brain I. Rini, Thomas Powles, Martin H. Voss, Laurence Albiges, and Eric Jonasch
- Abstract
Die kürzlich erfolgte Zulassung der Kombinationstherapien von Axitinib mit den Immuncheckpoint-Inhibitoren (ICI) Pembrolizumab oder Avelumab zur First-Line-Therapie des fortgeschrittenen Nierenzellkarzinoms macht Handlungsempfehlungen zur Differenzierung zwischen immunbezogenen Nebenwirkungen (adverse events, AEs), die durch ICI verursacht werden, und Axitinib-bezogenen AEs erforderlich, um die Therapie mit Axitinib-ICI-Kombinationen zu optimieren. Die hier vorgestellten Empfehlungen basieren auf einer systematischen kritischen Überprüfung der veröffentlichten Evidenzlage und einer Diskussion unter Experten auf diesem Gebiet sowie einer Umfrage mit dem Ziel, einen Expertenkonsens über spezifische Maßnahmen für das Therapiemanagement mit den Kombinationstherapien von Axitinib/Avelumab und Axitinib/Pembrolizumab zu erhalten. Die Experten identifizierten AE-Bereiche, bei denen ein spezielles Management während der Behandlung mit Axitinib-ICI-Kombinationstherapien notwendig ist und die von den aktuellen Empfehlungen nicht abgedeckt werden. Nebenwirkungen, die ein solches spezialisiertes Management erfordern, sind Diarrhoe, Lebertoxizität, Fatigue und kardiovaskuläre AEs. Die Triage zwischen immunsuppressiven und supportiven Maßnahmen ist ein zentrales Element des Therapiemanagements. Dieser neuartige Therapieansatz erfordert ein klinisches Monitoring und Erfahrungen mit beiden Wirkstoffklassen. In der vorliegenden Arbeit liegt der Schwerpunkt auf AEs, die Überschneidungen zwischen der Axitinib- und der ICI-Therapie aufweisen. Unsere Empfehlungen beziehen sich auf das Management von AEs, die unter der Behandlung mit Axitinib-ICI-Kombinationen auftreten, und haben zum Ziel, die Sicherheit dieser Therapien zu verbessern.
- Published
- 2020
- Full Text
- View/download PDF
104. Comprehensive Genomic Analysis of Metastatic Non–Clear-Cell Renal Cell Carcinoma to Identify Therapeutic Targets
- Author
-
Darren R. Feldman, David M. Hyman, Maria I. Carlo, Almedina Redzematovic, Debyani Chakravarty, Martin H. Voss, Yasser Ged, Marc Ladanyi, Ahmet Zehir, Nabeela Khan, Chung-Han Lee, Devyn Taylor Coskey, Robert J. Motzer, A. Ari Hakimi, Ying-Bei Chen, Sujata Patil, Mark E. Robson, and Jianjiong Gao
- Subjects
0301 basic medicine ,Cancer Research ,business.industry ,Cell ,medicine.disease ,03 medical and health sciences ,Clear cell renal cell carcinoma ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Oncology ,Renal cell carcinoma ,030220 oncology & carcinogenesis ,medicine ,Cancer research ,Original Report ,business - Abstract
PURPOSE Non–clear-cell renal cell carcinoma (nccRCC) encompasses approximately 20% of renal cell carcinomas and includes subtypes that vary in clinical and molecular biology. Compared with clear cell renal cell carcinoma, nccRCC demonstrates limited sensitivity to conventional vascular endothelial growth factor– and mammalian target of rapamycin–directed agents, indicating a need for better therapies. Characterizing the genomic landscape of metastatic nccRCC variants may help define novel therapeutic strategies. PATIENTS AND METHODS We retrospectively analyzed tumor tissue from patients with metastatic nccRCC who consented to genomic analysis of their tumor and germline DNA. A hybridization capture–based assay was used to identify single nucleotide variants and small insertions and deletions across more than 340 cancer-associated genes with germline comparison. Clinical actionability of somatic mutations was assessed using OncoKB levels of evidence. Microsatellite instability (MSI) in the tumor was investigated. RESULTS Of 116 patients included in the analysis, 57 (49%) presented with de novo metastatic disease, and 59 (51%) presented with localized disease that later metastasized. Subtype classifications included unclassified (n = 41; 35%), papillary (n = 26; 22%), chromophobe (n = 17; 15%), translocation associated (n = 13; 11%), and other (n = 19; 16%). Of all tumors, 15 (13%) had putative driver somatic alterations amenable to targeted therapies, including alterations in MET, TSC1/2, and an ALK translocation. Of 45 patients who had germline testing, 11 (24%) harbored mutations, seven of which could potentially guide therapy. Of 115 available tumors for analysis, two (1.7%) had high and six (5%) had intermediate MSI status. CONCLUSION The mutation profiles of metastatic nccRCC vary by subtype. Comprehensive analysis of somatic mutations, germline mutations, and MSI, interpreted via an annotated precision oncology knowledge base, identified potentially targetable alterations in 22% of patients, which merits additional investigation.
- Published
- 2019
- Full Text
- View/download PDF
105. Tumor Microenvironment Dynamics in Clear-Cell Renal Cell Carcinoma
- Author
-
A. Ari Hakimi, Martin H. Voss, Ritesh Kotecha, and Lynda Vuong
- Subjects
0301 basic medicine ,Stromal cell ,Angiogenesis ,T-Lymphocytes ,Article ,03 medical and health sciences ,Lymphocytes, Tumor-Infiltrating ,0302 clinical medicine ,Immune system ,Renal cell carcinoma ,Biomarkers, Tumor ,Tumor Microenvironment ,Animals ,Humans ,Medicine ,Molecular Targeted Therapy ,Carcinoma, Renal Cell ,Tumor microenvironment ,business.industry ,medicine.disease ,Combined Modality Therapy ,Kidney Neoplasms ,Immune checkpoint ,Clear cell renal cell carcinoma ,030104 developmental biology ,Oncology ,Drug development ,030220 oncology & carcinogenesis ,Cancer research ,business - Abstract
Renal cell carcinoma stands out as one of the most immune-infiltrated tumors in pan-cancer comparisons. Features of the tumor microenvironment heavily affect disease biology and may affect responses to systemic therapy. With evolving frontline options in the metastatic setting, several immune checkpoint blockade regimens have emerged as efficacious, and there is growing interest in characterizing features of tumor biology that can reproducibly prognosticate patients and/or predict the likelihood of their deriving therapeutic benefit. Herein, we review pertinent characteristics of the tumor microenvironment with dedicated attention to candidate prognostic and predictive signatures as well as possible targets for future drug development.Significance:Tumor microenvironment features broadly characterizing angiogenesis and inflammatory signatures have shown striking differences in response to immune checkpoint blockade and antiangiogenic agents. Integration of stromal and immune biomarkers may hence produce predictive and prognostic signatures to guide management with existing regimens as well as future drug development.
- Published
- 2019
- Full Text
- View/download PDF
106. Characterization and Impact of TERT Promoter Region Mutations on Clinical Outcome in Renal Cell Carcinoma
- Author
-
Brandon J. Manley, Paul Russo, Darren R. Feldman, Martin H. Voss, Mazyar Ghanaat, Jozefina Casuscelli, Ed Reznik, Jonathan A. Coleman, Maria E. Arcila, Christian G. Stief, A. Ari Hakimi, Victor E. Reuter, Robert J. Motzer, Mahyar Kashan, James J. Hsieh, Daniel M. Tennenbaum, Maria I. Carlo, Almedina Redzematovic, Emily C. Zabor, Ying-Bei Chen, and Maria F. Becerra
- Subjects
Adult ,Male ,0301 basic medicine ,Oncology ,medicine.medical_specialty ,Urology ,medicine.disease_cause ,Bioinformatics ,Article ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,Renal cell carcinoma ,Internal medicine ,medicine ,Humans ,Promoter Regions, Genetic ,Carcinoma, Renal Cell ,Telomerase ,Aged ,Retrospective Studies ,Aged, 80 and over ,Kidney ,business.industry ,Hazard ratio ,Middle Aged ,medicine.disease ,Primary tumor ,Kidney Neoplasms ,Survival Rate ,Clear cell renal cell carcinoma ,Exact test ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Mutation ,Female ,Carcinogenesis ,business ,Clear cell - Abstract
Background Mutations in the promoter region of the TERT gene have been detected in a variety of cancers. These mutations can potentially lead to unlimited cell divisions and result in poor clinical prognosis. Objective To determine the role and relevance of TERT promoter region mutations in both clear cell (ccRCC) and non–clear cell (nccRCC) renal cell carcinoma using ultra-deep and whole-genome sequencing methods on primary tumor samples. Design, setting, and participants DNA from 281 kidney tumors (147 ccRCC and 134 nccRCC) was sequenced between 2013 and 2015, and clinical outcomes for these patients from a single institution were retrospectively analyzed. Outcome measurements and statistical analysis Differences in patient characteristics and mutational status were tested using Fisher's exact test for categorical variables and the Wilcoxon rank sum test for continuous variables. Survival times were estimated using the Kaplan-Meier method and differences were tested using the log-rank test. Results and limitations TERT mutations occurred in 12.2% of ccRCC and 10.4% of nccRCC cases. In >80% of the cases, mutations were located at C228T and were found to co-occur only rarely with other relevant RCC driver genes. The median follow-up among survivors overall was 2.5 yr (range 0.1–18.3). TERT promoter mutations were significantly associated with cancer-specific survival in ccRCC (hazard ratio 2.68, 95% confidence interval 1.19–6.01; p =0.013). In nccRCC, TERT mutations were significantly associated with larger tumors and metastatic development. Assessment of further relevant clinical associations was precluded in the nccRCC group by the heterogeneous and small sample size. Conclusions Our data suggests that TERT mutational status reflects a distinct pathogenesis with an aggressive disease course in RCC. Stratifying patients with this unique tumorigenesis that leads to poor clinical outcomes could be a putative target for novel therapeutics. Patient summary We show a previously unrecognized frequency of TERT promoter mutations in both clear cell and non–clear cell renal cell carcinoma. TERT promoter mutations were associated with some worse outcomes in patients with clear cell renal cell carcinoma.
- Published
- 2019
- Full Text
- View/download PDF
107. Outcomes of patients with metastatic clear-cell renal cell carcinoma treated with second-line VEGFR-TKI after first-line immune checkpoint inhibitors
- Author
-
Ritesh Kotecha, Paul G. Corn, Jianjun Gao, Joshua Chaim, Amado J. Zurita, Lianchun Xiao, Nizar M. Tannir, Matthew T. Campbell, Emily Lemke, Martin H. Voss, Pavlos Msaouel, Anuradha Chandramohan, Robert J. Motzer, Jennifer Wang, Eric Jonasch, Amishi Yogesh Shah, and Padmanee Sharma
- Subjects
Male ,Vascular Endothelial Growth Factor A ,0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Bevacizumab ,Article ,Pazopanib ,03 medical and health sciences ,0302 clinical medicine ,Renal cell carcinoma ,Atezolizumab ,Internal medicine ,Humans ,Medicine ,Carcinoma, Renal Cell ,Protein Kinase Inhibitors ,Retrospective Studies ,business.industry ,Sunitinib ,Middle Aged ,medicine.disease ,Survival Analysis ,Kidney Neoplasms ,Clear cell renal cell carcinoma ,Treatment Outcome ,030104 developmental biology ,030220 oncology & carcinogenesis ,Female ,Immunotherapy ,Nivolumab ,business ,Progressive disease ,medicine.drug - Abstract
Background Immune checkpoint inhibitors (ICIs) are being increasingly utilised in the front-line (1L) setting of metastatic clear-cell renal cell carcinoma (mccRCC). Limited data exist on responses and survival on second-line (2L) vascular endothelial growth factor–receptor tyrosine kinase inhibitor (VEGFR-TKI) therapy after 1L ICI therapy. Patients and methods This is a retrospective study of mccRCC patients treated with 2L VEGFR-TKI after progressive disease (PD) with 1L ICI. Patients were treated at MD Anderson Cancer Center or Memorial Sloan Kettering Cancer Center between December 2015 and February 2018. Objective response was assessed by blinded radiologists' review using Response Evaluation Criteria in Solid Tumours v1.1. Descriptive statistics and Kaplan–Meier method were used. Results Seventy patients were included in the analysis. Median age at mccRCC diagnosis was 59 years; 8 patients (11%) had international metastatic database consortium favourable-risk disease, 48 (69%) had intermediate-risk disease and 14 (20%) had poor-risk disease. As 1L therapy, 12 patients (17%) received anti–programmed death ligand-1 (PD-(L)1) monotherapy with nivolumab or atezolizumab, 33 (47%) received nivolumab plus ipilimumab and 25 (36%) received combination anti–PD-(L)1 plus bevacizumab. 2L TKI therapies included pazopanib, sunitinib, axitinib and cabozantinib. On 2L TKI therapy, one patient (1.5%) achieved a complete response, 27 patients (39.7%) a partial response and 36 patients (52.9%) stable disease. Median progression-free survival (mPFS) was 13.2 months (95% confidence interval: 10.1, NA). Forty-five percent of subjects required a dose reduction, and twenty-seven percent of patients discontinued treatment because of toxicity. Conclusions In this retrospective study of patients with mccRCC receiving 2L TKI monotherapy after 1L ICI, we observed 2L antitumour activity and tolerance comparable to historical data for 1L TKI.
- Published
- 2019
- Full Text
- View/download PDF
108. A Phase I, Open-Label, Multicenter, Dose-escalation Study of the Oral Selective FGFR Inhibitor Debio 1347 in Patients with Advanced Solid Tumors Harboring FGFR Gene Alterations
- Author
-
Claudio Zanna, Cinta Hierro, A. John Iafrate, Josep Tabernero, Rebecca S. Heist, Nobuya Ishii, Funda Meric-Bernstam, Valerie Nicolas-Metral, James M. Cleary, Anna Pokorska-Bocci, Keith T. Flaherty, Martin H. Voss, Yulia Kirpicheva, Anne Vaslin Chessex, Leena Gandhi, José Baselga, Youyou Hu, Filip Janku, and Darrell R. Borger
- Subjects
0301 basic medicine ,Cancer Research ,medicine.medical_specialty ,business.industry ,medicine.disease ,Gastroenterology ,03 medical and health sciences ,Hyperphosphatemia ,030104 developmental biology ,0302 clinical medicine ,Oncology ,Tolerability ,Pharmacokinetics ,030220 oncology & carcinogenesis ,Pharmacodynamics ,Internal medicine ,Toxicity ,medicine ,FGFR Inhibitor Debio 1347 ,Adverse effect ,business ,Stomatitis - Abstract
Purpose: To investigate tolerability, efficacy, and pharmacokinetics/pharmacodynamics of Debio 1347, a selective FGFR inhibitor. Patients and Methods: This was a first-in-human, multicenter, open-label study in patients with advanced solid tumors harboring FGFR1–3 gene alterations. Eligible patients received oral Debio 1347 at escalating doses once daily until disease progression or intolerable toxicity. Dose-limiting toxicities (DLT) were evaluated during the first 4 weeks on treatment, pharmacokinetics/pharmacodynamics postfirst dose and after 4 weeks. Results: A total of 71 patients were screened and 58 treated with Debio 1347 at doses from 10 to 150 mg/day. Predominant tumor types were breast and biliary duct cancer, most common gene alterations were FGFR1 amplifications (40%) and mutations in FGFR2 (12%) and FGFR3 (17%); 12 patients (21%) showed FGFR fusions. Five patients at three dose levels had six DLTs (dry mouth/eyes, hyperamylasemia, hypercalcemia, hyperbilirubinemia, hyperphosphatemia, and stomatitis). The maximum tolerated dose was not reached, but dermatologic toxicity became sometimes dose limiting beyond the DLT period at ≥80 mg/day. Adverse events required dose modifications in 52% of patients, mostly due to dose-dependent, asymptomatic hyperphosphatemia (22%). RECIST responses were seen across tumor types and mechanisms of FGFR activation. Six patients, 3 with FGFR fusions, demonstrated partial responses, 10 additional patients' tumor size regressions of ≤30%. Plasma half-life was 11.5 hours. Serum phosphate increased with Debio 1347 plasma levels and confirmed target engagement at doses ≥60 mg/day. Conclusions: Preliminary efficacy was encouraging and tolerability acceptable up to 80 mg/day, which is now used in an extension part of the study.
- Published
- 2019
- Full Text
- View/download PDF
109. Treatment-free Survival after Immune Checkpoint Inhibitor Therapy versus Targeted Therapy for Advanced Renal Cell Carcinoma: 42-Month Results of the CheckMate 214 Trial
- Author
-
Brian Stwalley, Chung-Han Lee, Yoshihiko Tomita, Bernard Escudier, Hans J. Hammers, David F. McDermott, Thomas Powles, Martin H. Voss, Elizabeth R. Plimack, Viviana Del Tejo, Toni K. Choueiri, Meredith M. Regan, Michael B. Atkins, Laurence Albiges, Lillian Werner, Robert J. Motzer, Opeyemi Jegede, Brian I. Rini, Charlene Mantia, Nizar M. Tannir, and S. Huo
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Randomization ,medicine.medical_treatment ,Ipilimumab ,Article ,Targeted therapy ,Renal cell carcinoma ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Sunitinib ,Humans ,Adverse effect ,Carcinoma, Renal Cell ,Immune Checkpoint Inhibitors ,business.industry ,medicine.disease ,Kidney Neoplasms ,Toxicity ,Nivolumab ,business ,medicine.drug - Abstract
Purpose: Patients discontinuing immuno-oncology regimens may experience periods of disease control without need for ongoing anticancer therapy, but toxicity may persist. We describe treatment-free survival (TFS), with and without toxicity. Patients and Methods: Data were analyzed from the randomized phase III CheckMate 214 trial of nivolumab plus ipilimumab (n = 550) versus sunitinib (n = 546) for treatment-naïve, advanced renal cell carcinoma (aRCC). TFS was estimated by the 42-month restricted mean times defined by the area between Kaplan–Meier curves for two time-to-event endpoints defined from randomization: time to protocol therapy cessation and time to subsequent systemic therapy initiation or death. TFS was subdivided as TFS with and without toxicity by counting days with ≥1 grade ≥3 treatment-related adverse event (TRAE). Results: At 42 months since randomization, 52% of nivolumab plus ipilimumab and 39% of sunitinib intermediate/poor-risk patients were alive; 18% and 5% surviving treatment-free, respectively. Among favorable-risk patients, 70% and 73% of nivolumab plus ipilimumab and sunitinib patients were alive; 20% and 9% treatment-free. Over the 42-month period, mean TFS was over twice as long after nivolumab plus ipilimumab than sunitinib for intermediate/poor-risk (6.9 vs. 3.1 months) and three times as long for favorable-risk patients (11.0 vs. 3.7 months). Mean TFS with grade ≥3 TRAEs was a small proportion of time for both treatments (0.6 vs. 0.3 months after nivolumab plus ipilimumab vs. sunitinib for intermediate/poor-risk, and 0.9 vs. 0.3 months for favorable-risk patients). Conclusions: Patients initiating first-line nivolumab plus ipilimumab for aRCC spent more survival time treatment-free without toxicity versus those on sunitinib, regardless of risk group.
- Published
- 2021
110. Genomic characterization of metastatic patterns from prospective clinical sequencing of 25,000 patients
- Author
-
Charles M. Rudin, Brooke Mastrogiacomo, Bastien Nguyen, Julio Garcia-Aguilar, Ahmet Zehir, Christopher J. Fong, Ramyasree Madupuri, Eric Rios-Doria, Joan Massagué, Rona Yaeger, Nadeem R. Abu-Rustum, Carol Aghajanian, Ed Reznik, Eileen M. O'Reilly, Nikolaus Schultz, Subhiksha Nandakumar, Michael A. Postow, Christine A. lacobuzio-Donahue, Luc G. T. Morris, Darren R. Feldman, Robert J. Motzer, Pedram Razavi, Ghassan K. Abou-Alfa, Mithat Gonen, William R. Jarnagin, Michael J. Morris, Vivian E. Strong, Sarat Chandarlapaty, Michael F. Berger, William D. Tap, Andrea Cercek, Nitya Raj, Alan L. Ho, Min Yuen Teo, James J. Harding, Adrienne Boire, Lora H. Ellenson, Bernard H. Bochner, Sohrab P. Shah, Anna M. Varghese, Craig M. Bielski, Wassim Abida, Jonathan E. Rosenberg, David B. Solit, Anton Safonov, Ritika Kundra, Henry Walch, Renzo G. DiNatale, Martin H. Voss, Francisco Sanchez-Vega, David R. Jones, Alexander N. Shoushtari, Shaleigh A. Smith, Samuel Singer, Mark E. Robson, Karuna Ganesh, Anisha Luthra, Debyani Chakravarty, Jianjiong Gao, Britta Weigelt, Dmitriy Zamarin, A.A. Hakimi, Yelena Y. Janjigian, Daniela Molena, Marc Ladanyi, Walid K. Chatila, Gregory J. Riely, Jorge S. Reis-Filho, Samuel F. Bakhoum, Gopakumar Iyer, Marjorie G. Zauderer, Paul Russo, Anuradha Gopalan, and Ping Chi
- Subjects
Lung ,Somatic cell ,business.industry ,Cancer ,Disease ,medicine.disease ,Metastasis ,medicine.anatomical_structure ,Chromosome instability ,Cohort ,medicine ,Cancer research ,Adenocarcinoma ,business - Abstract
Progression to metastatic disease remains the main cause of cancer death. Yet, the underlying genomic mechanisms driving metastasis remain largely unknown. Here, we present MSK-MET, an integrated pan-cancer cohort of tumor genomic and clinical outcome data from more than 25,000 patients. We analyzed this dataset to identify associations between tumor genomic alterations and patterns of metastatic dissemination across 50 tumor types. We found that chromosomal instability is strongly correlated with metastatic burden in some tumor types, including prostate adenocarcinoma, lung adenocarcinoma and HR-positive breast ductal carcinoma, but not in others, such as colorectal adenocarcinoma, pancreatic adenocarcinoma and high-grade serous ovarian cancer. We also identified specific somatic alterations associated with increased metastatic burden and specific routes of metastatic spread. Our data offer a unique resource for the investigation of the biological basis for metastatic spread and highlight the crucial role of chromosomal instability in cancer progression.
- Published
- 2021
- Full Text
- View/download PDF
111. Single cell sequencing links multiregional immune landscapes and tissue-resident T cells in ccRCC to tumor topology and therapy efficacy
- Author
-
Mahdi Golkaram, Timothy A. Chan, Christina S. Leslie, Victor E. Reuter, Raakhee Vijayaraghavan, Fengshen Kuo, Paul Russo, Kyrollis Attalla, Maria I. Carlo, Traci Pawlowski, Tanaya A. Purohit, Martin H. Voss, Chirag Krishna, Jonathan A. Coleman, Briana G. Nixon, Ming Liu, Stanley Weng, Raghvendra M. Srivastava, Sounak Gupta, Ming O. Li, Robert J. Motzer, Chad M. Vanderbilt, Michael L. Salmans, A. Ari Hakimi, Kyle A. Blum, Renzo G. DiNatale, Shile Zhang, Diego Chowell, Ying-Bei Chen, Vladimir Makarov, Lynda Vuong, Emily R. Kansler, and Li Liu
- Subjects
0301 basic medicine ,Cancer Research ,medicine.medical_treatment ,T-Lymphocytes ,Cell ,Programmed Cell Death 1 Receptor ,Receptors, Antigen, T-Cell ,Biology ,CD8-Positive T-Lymphocytes ,Lymphocyte Activation ,Article ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,medicine ,Humans ,Lymph node ,Carcinoma, Renal Cell ,Macrophages ,T-cell receptor ,Immunotherapy ,Immune checkpoint ,Kidney Neoplasms ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,Single cell sequencing ,030220 oncology & carcinogenesis ,Cancer research ,Disease Progression ,Clear cell - Abstract
Summary Clear cell renal cell carcinomas (ccRCCs) are highly immune infiltrated, but the effect of immune heterogeneity on clinical outcome in ccRCC has not been fully characterized. Here we perform paired single-cell RNA (scRNA) and T cell receptor (TCR) sequencing of 167,283 cells from multiple tumor regions, lymph node, normal kidney, and peripheral blood of two immune checkpoint blockade (ICB)-naive and four ICB-treated patients to map the ccRCC immune landscape. We detect extensive heterogeneity within and between patients, with enrichment of CD8A+ tissue-resident T cells in a patient responsive to ICB and tumor-associated macrophages (TAMs) in a resistant patient. A TCR trajectory framework suggests distinct T cell differentiation pathways between patients responding and resistant to ICB. Finally, scRNA-derived signatures of tissue-resident T cells and TAMs are associated with response to ICB and targeted therapies across multiple independent cohorts. Our study establishes a multimodal interrogation of the cellular programs underlying therapeutic efficacy in ccRCC.
- Published
- 2021
112. Characterization of FH-deficient renal cell carcinoma
- Author
-
Ying-Bei Chen, Ritesh Kotecha, Robert J. Motzer, Jack Patrick Gleeson, Mark Zucker, Andrea Knezevic, Martin H. Voss, Jonathan A. Coleman, Maria I. Carlo, Samuel Murray, A. Ari Hakimi, Sujata Patil, Ines Nikolovski, Darren R. Feldman, Ed Reznik, Renzo G. DiNatale, Zsofia K. Stadler, Natalie Shapnik, Yasser Ged, Paul Russo, and Chung-Han Lee
- Subjects
0301 basic medicine ,Adult ,Male ,Cancer Research ,DNA Copy Number Variations ,Somatic cell ,medicine.medical_treatment ,Urology ,MEDLINE ,medicine.disease_cause ,urologic and male genital diseases ,Germline ,Article ,Fumarate Hydratase ,03 medical and health sciences ,Young Adult ,0302 clinical medicine ,Text mining ,Renal cell carcinoma ,Antineoplastic Combined Chemotherapy Protocols ,Biomarkers, Tumor ,medicine ,Humans ,Carcinoma, Renal Cell ,PI3K/AKT/mTOR pathway ,Alleles ,Germ-Line Mutation ,Aged ,Neoplasm Staging ,Retrospective Studies ,Mutation ,business.industry ,Computational Biology ,Immunotherapy ,Middle Aged ,Prognosis ,medicine.disease ,Immunohistochemistry ,030104 developmental biology ,Treatment Outcome ,Oncology ,030220 oncology & carcinogenesis ,Fumarase ,Cancer research ,Female ,Neoplasm Grading ,business - Abstract
Purpose: Fumarate hydratase–deficient renal cell carcinoma (FH-RCC) is a rare, aggressive form of RCC associated with hereditary leiomyomatosis and RCC syndrome. Evidence for systemic therapy efficacy is lacking. Experimental Design: We studied clinical and genomic characteristics of FH-RCC, including response [objective response rate (ORR)] to systemic therapies and next-generation sequencing (NGS). Patients with metastatic FH-RCC, defined by presence of pathogenic germline or somatic FH mutation plus IHC evidence of FH loss, were included. Results: A total of 28 of 32 included patients (median age 46; range, 20–74; M:F, 20:12) underwent germline testing; 23 (82%) harbored a pathogenic FH germline variant. Five (16%) were negative for germline FH mutations; all had biallelic somatic FH loss. Somatic NGS (31/32 patients) revealed co-occurring NF2 mutation most frequently (n = 5). Compared with clear-cell RCC, FH-RCC had a lower mutation count (median 2 vs. 4; P < 0.001) but higher fraction of genome altered (18.7% vs. 10.3%; P = 0.001). A total of 26 patients were evaluable for response to systemic therapy: mTOR/VEGF combination (n = 18, ORR 44%), VEGF monotherapy (n = 15, ORR 20%), checkpoint inhibitor therapy (n = 8, ORR 0%), and mTOR monotherapy (n = 4, ORR 0%). No complete responses were seen. Median overall and progression-free survival were 21.9 months [95% confidence interval (CI): 14.3–33.8] and 8.7 months (95% CI: 4.8–12.3), respectively. Conclusions: Although most FH-RCC tumors are due to germline FH alterations, a significant portion result from biallelic somatic FH loss. Both somatic and germline FH-RCC have similar molecular characteristics, with NF2 mutations, low tumor mutational burden, and high fraction of genome altered. Although immunotherapy alone produced no objective responses, combination mTOR/VEGF therapy showed encouraging results.
- Published
- 2021
113. Evolving biological associations of upfront cytoreductive nephrectomy in metastatic renal cell carcinoma
- Author
-
Martin H. Voss, Andrew W. Silagy, Kyrollis Attalla, Arturo Holmes, Robert J. Motzer, Jonathan A. Coleman, Roy Mano, Paul Russo, Kate Weiss, Nirmish Singla, A. Ari Hakimi, Ritesh Kotecha, Renzo G. DiNatale, Sujata Patil, and Stanley Weng
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Thrombocytosis ,Performance status ,Anemia ,business.industry ,Hazard ratio ,Cytoreduction Surgical Procedures ,medicine.disease ,Nephrectomy ,Neutrophilia ,Confidence interval ,Kidney Neoplasms ,Renal cell carcinoma ,Internal medicine ,medicine ,Humans ,medicine.symptom ,Risk factor ,business ,Carcinoma, Renal Cell ,Retrospective Studies - Abstract
Background Systemic responses to cytoreductive nephrectomy (CN) in the management of metastatic renal cell carcinoma (mRCC) are variable and difficult to anticipate. The authors aimed to determine the association of CN with modifiable International Metastatic Renal Cell Carcinoma Database Consortium (IMDC) risk factors and oncological outcomes. Methods Consecutive patients with mRCC referred for potential CN (2009-2019) were reviewed. The primary outcome was overall survival (OS); variables of interest included undergoing CN and the baseline number of modifiable IMDC risk factors (anemia, hypercalcemia, neutrophilia, thrombocytosis, and reduced performance status). For operative cases, the authors evaluated the effects of IMDC risk factor dynamics, measured 6 weeks and 6 months after CN, on OS and postoperative treatment disposition. Results Of 245 treatment-naive patients with mRCC referred for CN, 177 (72%) proceeded to surgery. The CN cases had fewer modifiable IMDC risk factors (P = .003), including none in 71 of 177 patients (40.1%); fewer metastases (P = .011); and higher proportions of clear cell histology (P = .012). In a multivariable analysis, surgical selection, number of IMDC risk factors, metastatic focality, and histology were associated with OS. Total risk factors changed for 53.8% and 57.2% of the patients from the preoperative period to 6 weeks and 6 months after CN, respectively. Adjusted for preoperative IMDC risk scores, an increase in IMDC risk factors at 6 weeks and 6 months was associated with adverse OS (hazard ratio [HR], 1.57; 95% confidence interval [CI], 1.13-2.19; P = .007; HR, 2.52; 95% CI, 1.74-3.65; P Conclusions IMDC risk factors are dynamic clinical variables that can improve after upfront CN in select patients, and this suggests a systemic benefit of cytoreduction, which may confer clinically meaningful prognostic implications.
- Published
- 2021
114. Success and failure of additional immune modulators in steroid-refractory/resistant pneumonitis related to immune checkpoint blockade
- Author
-
Michael A. Postow, Matthew D. Hellmann, Hira Rizvi, Jason Beattie, Mohit Chawla, Allison Betof Warner, Adam J. Schoenfeld, Martin H. Voss, Margaret K. Callahan, Jia Luo, I-Hsin Lin, Paige Fuentes, and Neil J. Shah
- Subjects
Oncology ,Male ,Cancer Research ,medicine.medical_specialty ,Time Factors ,medicine.medical_treatment ,Immunology ,Drug Resistance ,03 medical and health sciences ,Immunomodulating Agents ,0302 clinical medicine ,Immune system ,Internal medicine ,medicine ,Immunology and Allergy ,Humans ,030212 general & internal medicine ,Adverse effect ,Immune Checkpoint Inhibitors ,RC254-282 ,Pneumonitis ,Aged ,Retrospective Studies ,Pharmacology ,Clinical/Translational Cancer Immunotherapy ,business.industry ,Cancer ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Immunosuppression ,Retrospective cohort study ,Immunotherapy ,Pneumonia ,Middle Aged ,medicine.disease ,Immune checkpoint ,Treatment Outcome ,inflammation ,030220 oncology & carcinogenesis ,Molecular Medicine ,Female ,New York City ,Steroids ,immunotherapy ,business - Abstract
BackgroundPneumonitis related to immune checkpoint blockade is uncommon but can be severe, fatal or chronic. Steroids are first-line treatment, however, some patients are refractory or become resistant to steroids. Like many immune-related adverse events, little is known regarding the outcomes and optimal management of patients in whom steroids are ineffective.MethodsWe performed a single-center retrospective cohort study at a high-volume tertiary cancer center to evaluate the clinical course, management strategies and outcomes of patients treated for immune checkpoint pneumonitis with immune modulatory medications in addition to systemic steroids. Pharmacy records were queried for patients treated with both immune checkpoint blockade and receipt of additional immune modulators. Records were then manually reviewed to identify patients who received the additional immune modulators for immune checkpoint pneumonitis.ResultsFrom 2013 to 2020, we identified 26 patients treated for immune checkpoint pneumonitis with additional immune modulators in addition to steroids. Twelve patients (46%) were steroid-refractory and 14 (54%) were steroid-resistant. Pneumonitis severity included grade 2 (42%) or grade 3–4 (58%). Additional immune modulation consisted of tumor necrosis factor-alpha inhibitor (77%) and/or mycophenolate (23%). Durable improvement in pneumonitis following initiation of additional immune modulators occurred in 10 patients (38%), including three patients (12%) in whom pneumonitis resolved and all immunosuppressants ceased. The rate of 90-day all-cause mortality/hospice referral was 50%. At last follow-up, mortality attributable to pneumonitis was 23%. In addition to mortality from pneumonitis and cancer, 3 patients (12%) died due to infections possibly associated with immunosuppression.ConclusionsSteroid-refractory or -resistant immune checkpoint pneumonitis is uncommon but associated with significant morbidity and mortality. Additional immunomodulators can yield durable improvement, attained in over one third of patients. An improved understanding of the underlying biology of immune-related pneumonitis will be crucial to guide more precise and effective treatment strategies in the future.
- Published
- 2021
115. A robust, trial-agnostic gene signature for response to tyrosine kinase inhibition in ccRRCC
- Author
-
Sari Safaa Khaleel, Minwei Liu, Fengshen Kuo, Ritesh Kotecha, Chung-Han Lee, Martin H Voss, Robert J. Motzer, A. Ari Hakimi, and Eduard Reznik
- Subjects
Cancer Research ,Oncology - Abstract
4558 Background: Combination systemic therapies (ST) including immunotherapy (IO) and Tyrosine Kinase Inhibitors (TKI) have become the standard of care for management of metastatic clear cell carcinoma (ccRCC). Several expression (GE) signatures have been reported to predict TKI response with the aim of identifying the optimal ST regimen (IO/IO or IO/TKI) per patient. However, these gene signatures were developed and evaluated using data from individual trials with clinically distinct patient cohorts, fundamentally impeding their general application to diverse and heterogeneous patient populations. We therefore sought to identify genes consistently and directly associated with progression-free survival (PFS) with TKI ST across several trials. Methods: We applied a stratified, weighted concordance model to identify genes consistently demonstrating statistically significant association with PFS in the sunitinib TKI arm in four large randomized controlled trials (RCTs) of ST in ST-naïve ccRCC patients: COMPARZ (Motzer et al, 2013), Checkmate 214 (Motzer et al, 2018), IMmotion 151 (Rini et al, 2019), and Javelin 101 (Motzer et al, 2019). Concordant genes with Benjamini-Hochberg adjusted p-value (pAdj) ≤ 0.01 were selected. Of these, genes significantly associated with PFS on Cox proportional hazard regression (pAdj ≤ 0.05) in at least 3 of the 4 datasets were selected. Ontologic process and pathway enrichment analyses (OPPEA) against several ontologic databases were peformed using Metascape (Zhou et al, 2019). Results: We identified 2,794 statistically significant genes demonstrating concordant association with PFS across all 4 datasets (1087 patients); 71 genes were significantly associated with PFS on univariate post-hoc analysis in ≥3 of the datasets. OPPEA of these results identified significant enrichment in genes associated with cell cycle, cell division, and VEGFA-VEGFR2 pathways (Table 1). Although 5 genes in our set overlapped with published angiogenesis GES, the majority of these genes constitute entirely novel transcriptomic correlates of response to sunitinib. Conclusions: We developed a robust meta-gene signature consisting of genes directly and consistently associated with TKI response using data from several clinical trials. This gene signature is readily applicable to heterogeneous patient populations treated with anti-VEGF therapies. [Table: see text]
- Published
- 2022
- Full Text
- View/download PDF
116. Molecular profile and clinical outcomes of renal cell carcinoma brain metastases treated with stereotactic radiosurgery
- Author
-
Jennifer Ma, Luke del Balzo, Sari Safaa Khaleel, Jessica Flynn, Zhigang Zhang, Martin H Voss, Benjamin Freeman, A. Ari Hakimi, Chung-Han Lee, Jordan Eichholz, Daniel W. Kelly, Jonathan T. Yang, Boris Mueller, Maria Isabel Carlo, Robert J. Motzer, Brandon S. Imber, Kathryn Beal, Nelson S. Moss, Ritesh Kotecha, and Luke Roy George Pike
- Subjects
Cancer Research ,Oncology - Abstract
4526 Background: Molecular profiles of renal cell carcinoma (RCC) tumors are associated with systemic treatment (ST) responses and clinical outcomes. However, the molecular profiles of RCC brain metastases (BM) and their correlation with ST response and clinical outcomes are not well characterized. Effective management of BM with locoregional therapies including stereotactic radiosurgery (SRS) is critical as ST advances have improved overall survival (OS). Therefore, we sought to identify the clinical and genomic features of RCC BM in a large cohort of patients treated with SRS. Methods: We performed an institutional retrospective analysis of RCC BM patients treated with SRS and evaluated corresponding genomic next generation sequencing (NGS) data via a targeted sequencing panel (MSK-IMPACT). A comparison cohort of all institutional patients with available NGS data was utilized to investigate genes enriched in our BM cohort using Fisher exact testing. Kaplan Meier analyses were performed for OS and intracranial progression-free survival (iPFS). Clinical factors and genes mutated in ≥ 10% of samples were assessed per patient using Cox proportional hazards models, and per individual BMs using clustered competing risks regression with a competing risk of death. Results: From 2010-2021, 91 RCC BM patients underwent SRS for 212 BMs, including 86% clear cell and 14% non-clear cell RCC. NGS data was available for 76 patients (84%), including 18 resected BMs, 26 extra-cranial metastatic lesions (EM), and 32 primary kidney tumors (Table 1). Median follow-up was 3.2 years with median OS of 21 months (m) and median iPFS of 7.8m. Karnofsky performance status ≥80 and extracranial disease control were significantly associated with improved OS on multivariable analyses (MVA; p=0.049 and 0.01, respectively). No clinical variables were significantly associated with iPFS on MVA. At the BM level, SETD2 alterations approached significance for improved iPFS (HR=0.35; 95%CI 0.11, 1.05; p=0.06). Enrichment in SMARCA4 alterations was seen in the BM cohort as compared to primary kidney and EM samples from patients without BM (17% vs 1% vs 2%, p
- Published
- 2022
- Full Text
- View/download PDF
117. Progression-free survival after second line of therapy (PFS-2) for metastatic clear cell renal cell carcinoma (ccRCC) in patients treated with first-line immunotherapy combinations
- Author
-
Kelly N. Fitzgerald, Cihan Duzgol, Andrea Knezevic, Natalie Shapnik, Ritesh Kotecha, David Henry Aggen, Maria Isabel Carlo, Neil J. Shah, Martin H Voss, Darren R. Feldman, Robert J. Motzer, and Chung-Han Lee
- Subjects
Cancer Research ,Oncology - Abstract
4536 Background: Front-line therapy with immunotherapy combinations is standard of care for metastatic ccRCC, with ipilimumab/nivolumab (IO/IO) and several combinations of a VEGFR-targeted tyrosine kinase inhibitor with a PD-1 inhibitor (TKI/IO) showing superior efficacy to TKI monotherapy. PFS-2 evaluates the ability to be salvaged by 2nd line therapy and is a surrogate for overall survival (OS). PFS-2 was compared in patients receiving 1st line IO/IO vs TKI/IO for metastatic ccRCC. Methods: A retrospective analysis was performed on patients with ccRCC treated at Memorial Sloan Kettering Cancer Center between 1/1/2014 and 12/30/2020, in cohorts defined by 1st line: IO/IO or TKI/IO. PFS-2 is defined as time from start of 1st line to progression on next therapy, or death. Patients without a PFS-2 event were censored at a prespecified cutoff date. Objective response rate to 1st (ORR1st) and 2nd (ORR2nd) line are compared with the Fisher’s exact test. OS, PFS-2, and time on therapy are estimated with the Kaplan-Meier method and compared with the log-rank test. Results: One hundred seventy-three patients received 1st line IO/IO (N = 90) or 1st line TKI/IO (N = 83); respectively, 52 and 40 patients had a PFS-2 event. 1st line TKI/IO regimens included: 34% axitinib/pembrolizumab, 29% lenvatinib/pembrolizumab, 25% axitinib/avelumab, 11% other. More IO/IO patients had brain metastases and intermediate/poor MSKCC risk category (respectively p = 0.007, p < 0.001). ORR1st and median months on 1st line were higher with TKI/IO vs IO/IO (65% vs 39%, p < 0.001; 16.1 vs 5.1, p < 0.001). ORR2nd was higher with IO/IO vs TKI/IO (47% vs 13%, p < 0.001), and median months on 2nd line was not significantly different (7.7 vs 7.1, p = 0.30). Median PFS-2 for TKI/IO was 44 months (95% CI: 27, 53) vs 23 months (95% CI: 16, 47) for IO/IO, p = 0.13. For TKI/IO and IO/IO groups, respective PFS-2 at 12 months was 86% (95% CI 77, 92) and 74% (95% CI 63, 82); PFS-2 at 36 months was 51% (95% CI 39, 63) and 42% (95% CI 30, 53). OS was not significantly different (p = 0.32; 3 year OS: IO/IO 60%, 95% CI 47, 71; TKI/IO 62%, 95% CI 49, 73). (Table) Conclusions: In patients receiving 1st line IO/IO or TKI/IO, ORR2nd was higher with IO/IO and median PFS-2 was numerically higher with TKI/IO, but no statistically significant difference in PFS-2 or OS was seen. These findings suggest that IO/IO and TKI/O are both acceptable 1st line treatment strategies in ccRCC. [Table: see text]
- Published
- 2022
- Full Text
- View/download PDF
118. STARLITE 2: Phase 2 study of nivolumab plus 177Lutetium-labeled anti-carbonic anhydrase IX (CAIX) monoclonal antibody girentuximab (177Lu-girentuximab) in patients (pts) with advanced clear cell renal cell carcinoma (ccRCC)
- Author
-
Darren R. Feldman, Robert J. Motzer, Andrea Knezevic, Chung-Han Lee, Martin H Voss, Serge K. Lyashchenko, Hijin Park, Steven M. Larson, and Neeta Pandit-Taskar
- Subjects
Cancer Research ,Oncology - Abstract
TPS752 Background: CAIX is a cell surface glycoprotein expressed in >90% of ccRCC but rarely in normal tissues, providing a target for imaging and therapeutic application. Radiolabeling the anti-CAIX monoclonal antibody girentuximab with 89Zr has shown promise as a novel PET tracer and labeling with 177Lu promise as a therapeutic agent in ccRCC (Muselaers, Eur Urol, 2016). Targeted delivery of radiation to ccRCC cells may prime the immune response by enhancing tumor antigen presentation, providing rationale for combining 177Lu-girentuximab with the anti-PD-1 antibody, nivolumab. This phase 2, open-label, single arm study (NCT05239533) is being conducted to evaluate 177Lu-girentuximab in combination with nivolumab in pts with previously treated ccRCC. Methods: Pts with biopsy-proven ccRCC, progressive disease after prior systemic therapy including ≥1 immunotherapy (IO) agent, adequate organ/marrow function, and ≥1 evaluable lesion by RECIST 1.1 that is also avid on 89Zr-girentuximab PET will be enrolled. There is no limit on number of prior lines of systemic therapy, but pts who stopped IO for immune toxicity are excluded. Treatment consists of 177Lu-girentuximab every 12-14 weeks for a maximum of 3 doses plus nivolumab 240mg every 2 weeks until progressive disease (PD) or unacceptable toxicity. Due to expected cumulative myelosuppression, each subsequent 177Lu-girentuximab dose to the same patient is reduced by 25% (dose 2 = 75% of dose 1; dose 3 = 75% of dose 2). Tumor imaging is performed every 12 weeks. Pts will be evaluated in a safety lead-in phase followed by an expansion phase. In the safety lead-in phase, the MTD of 177Lu-girentuximab in combination with nivolumab will be determined with a 3+3 design using a starting dose of 1804 MBq/m2 (75% of single agent MTD). For cohort 2, dose escalation to 2405 MBq/m2 (single agent MTD) or de-escalation to 1353 MBq/m2 will be based on dose-limiting toxicities. In the expansion phase, a Simon 2-stage optimal design is used to evaluate the primary endpoint of response rate by RECIST 1.1 within 24 weeks. With ≥1 response in the first Simon stage (n=10; includes pts treated at MTD in the safety lead-in phase), a second stage will open (n=19) for a total of 29 pts with ≥3 responses indicating the regimen worthy of further study. Secondary endpoints include PFS, OS, and toxicity including a continuous safety monitoring rule during expansion. Exploratory imaging with 89Zr-girentuximab PET is performed at baseline and before each 177Lu-girentuximab dose with results correlated with RECIST response on conventional imaging. In addition, whole body planar and SPECT imaging are performed after each 177Lu-girentuximab dose to evaluate distribution, lesion uptake and dosimetry of 177Lu-girentuximab. The trial is currently accruing to the safety lead-in phase. Clinical trial information: NCT05239533 .
- Published
- 2022
- Full Text
- View/download PDF
119. 717TiP Randomized, open-label, 3-arm phase III study comparing MK-1308A + lenvatinib and pembrolizumab (pembro) + belzutifan + lenvatinib versus pembro + lenvatinib as first-line (1L) treatment for advanced clear cell renal cell carcinoma (ccRCC)
- Author
-
Brian I. Rini, Rodolfo F. Perini, Elizabeth R. Plimack, Rachel Kloss Silverman, Karla Rodriguez-Lopez, Thomas Powles, Howard Gurney, Martin H. Voss, and Toni K. Choueiri
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,First line ,Hematology ,Pembrolizumab ,medicine.disease ,chemistry.chemical_compound ,Clear cell renal cell carcinoma ,chemistry ,Internal medicine ,Medicine ,Open label ,business ,Lenvatinib - Published
- 2021
- Full Text
- View/download PDF
120. Novel standards and emerging therapies for systemically treatment-naïve clear cell renal cell carcinoma - a rapidly changing landscape
- Author
-
Ritesh R. Kotecha and Martin H. Voss
- Abstract
The last decade has brought about major advances in systemic therapy for patients with advanced clear cell renal cell carcinoma. The introduction of angiogenesis targeting agents, immune checkpoint inhibitors, and combinations thereof has resulted in a multitude of therapeutic standards for patients with newly diagnosed advanced disease. With the rapid adoption and increasing number of available options for patients, selection amongst treatment regimens has become complex. Further, a new balance is also being sought to optimize treatment outcomes and limit treatment-related toxicities. With a rising bar against which novel therapeutics are being measured, the field looks toward an evolved understanding of tumor biology to help pave new ways forward for individualized therapy. Here, we review pivotal studies that led to regulatory approvals and ongoing clinical trials conducted in patients with systemically untreated clear cell renal cell carcinoma and provide perspective on how newly emerging data can be integrated into this rapidly changing landscape.
- Published
- 2021
- Full Text
- View/download PDF
121. Phase 1 study of mTORC1/2 inhibitor sapanisertib (TAK-228) in advanced solid tumours, with an expansion phase in renal, endometrial or bladder cancer
- Author
-
David Smith, Roberto Pili, Farhad Sedarati, Shadia I. Jalal, Andrés Cervantes, Martin H. Voss, Howard A. Burris, Rachel Neuwirth, Teresa Macarulla, Douglas V. Faller, Michael S. Gordon, Monica M. Mita, Vicky Makker, Igor Puzanov, Ding Wang, A. Enke, Brian I. Rini, Shubham Pant, Manish R. Patel, Yaping Shou, Institut Català de la Salut, [Voss MH, Makker V] Department of Medicine, 300 East 66th Street, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA. [Gordon MS] Oncology Research, HonorHealth Research Institute, 10510 N 92nd St Suite 200, Scottsdale, AZ 85258, USA. [Mita M] Department of Hematology and Oncology, Cedars-Sinai Medical Center, Samuel Oschin Comprehensive Cancer Institute, 8700 Beverly Blvd North Tower, Los Angeles, CA 90048, USA. [Rini B] Cleveland Clinic Foundation, Department of Solid Tumor Oncology, 9500 Euclid Avenue, Cleveland, OH 44195, USA. [Macarulla T] Servei d’Oncologia Mèdica, Vall d’Hebron Hospital Universitari, Barcelona, Spain. Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain, and Vall d'Hebron Barcelona Hospital Campus
- Subjects
Male ,Cancer Research ,Cancer therapy ,Other subheadings::Other subheadings::/drug therapy [Other subheadings] ,0302 clinical medicine ,Renal cell carcinoma ,Neoplasms ,Medicine ,Carcinoid tumour ,Aged, 80 and over ,0303 health sciences ,TOR Serine-Threonine Kinases ,Càncer - Tractament ,Middle Aged ,Kidney Neoplasms ,Oncology ,Tolerability ,030220 oncology & carcinogenesis ,técnicas de investigación::pruebas de toxicidad::dosis máxima tolerada [TÉCNICAS Y EQUIPOS ANALÍTICOS, DIAGNÓSTICOS Y TERAPÉUTICOS] ,Female ,Adult ,medicine.medical_specialty ,Maximum Tolerated Dose ,Urology ,Otros calificadores::Otros calificadores::/farmacoterapia [Otros calificadores] ,Antineoplastic Agents ,Urological cancer ,Article ,neoplasias [ENFERMEDADES] ,03 medical and health sciences ,Pharmacokinetics ,Carcinoma ,Humans ,Carcinoma, Renal Cell ,Protein Kinase Inhibitors ,Aged ,030304 developmental biology ,Gynaecological cancer ,Bladder cancer ,business.industry ,Endometrial cancer ,Investigative Techniques::Toxicity Tests::Maximum Tolerated Dose [ANALYTICAL, DIAGNOSTIC AND THERAPEUTIC TECHNIQUES, AND EQUIPMENT] ,medicine.disease ,Endometrial Neoplasms ,Neoplasms [DISEASES] ,Pyrimidines ,Urinary Bladder Neoplasms ,Pharmacodynamics ,Pyrazoles ,Medicaments - Administració ,business - Abstract
Background This Phase 1 dose-escalation/expansion study assessed safety/tolerability of sapanisertib, an oral, highly selective inhibitor of mTORC1/mTORC2, in advanced solid tumours. Methods Eligible patients received increasing sapanisertib doses once daily (QD; 31 patients), once weekly (QW; 30 patients), QD for 3 days on/4 days off QW (QD × 3dQW; 33 patients) or QD for 5 days on/2 days off QW (QD × 5dQW; 22 patients). In expansion cohorts, 82 patients with renal cell carcinoma (RCC), endometrial or bladder cancer received sapanisertib 5 mg QD (39 patients), 40 mg QW (26 patients) or 30 mg QW (17 patients). Results Maximum tolerated doses of sapanisertib were 6 mg QD, 40 mg QW, 9 mg QD × 3dQW and 7 mg QD × 5dQW. Frequent dose-limiting toxicities (DLTs) included hyperglycaemia, maculo-papular rash (QD), asthenia and stomatitis (QD × 3dQW/QD × 5dQW); expansion phase doses of 5 mg QD and 30 mg QW were selected based on tolerability beyond the DLT evaluation period. One patient with RCC achieved complete response; nine experienced partial responses (RCC: seven patients; carcinoid tumour/endometrial cancer: one patient each). Sapanisertib pharmacokinetics were time-linear and supported multiple dosing. Pharmacodynamic findings demonstrated treatment-related reductions in TORC1/2 biomarkers. Conclusions Sapanisertib demonstrated a manageable safety profile, with preliminary antitumour activity observed in RCC and endometrial cancer. Clinical trial registration ClinicalTrials.gov, NCT01058707.
- Published
- 2020
122. Prevalence and Landscape of Actionable Genomic Alterations in Renal Cell Carcinoma
- Author
-
Paul Russo, A. Ari Hakimi, Nikolaus Schultz, Chung-Han Lee, Victor E. Reuter, Ed Reznik, Renzo G. DiNatale, Kyrollis Attalla, Phillip M Rappold, Timothy A. Chan, Francisco Sanchez-Vega, Robert J. Motzer, Stanley Weng, Martin H. Voss, Jeremy C. Durack, Andrew W. Silagy, Stephen B. Solomon, Jonathan A. Coleman, Maria I. Carlo, and Christopher J. Fong
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Genome ,business.industry ,Cancer ,Microsatellite instability ,High-Throughput Nucleotide Sequencing ,Disease ,medicine.disease ,Kidney Neoplasms ,Article ,Renal cell carcinoma ,Localized disease ,Internal medicine ,Cohort ,Mutation ,medicine ,Humans ,DNA mismatch repair ,Stage (cooking) ,business ,Carcinoma, Renal Cell - Abstract
Purpose: We report our experience with next-generation sequencing to characterize the landscape of actionable genomic alterations in renal cell carcinoma (RCC). Experimental Design: A query of our institutional clinical sequencing database (MSK-IMPACT) was performed that included tumor samples from 38,468 individuals across all cancer types. Somatic variations were annotated using a precision knowledge database (OncoKB) and the available clinical data stratified by level of evidence. Alterations associated with response to immune-checkpoint blockade (ICB) were analyzed separately; these included DNA mismatch repair (MMR) gene alterations, tumor mutational burden (TMB), and microsatellite instability (MSI). Data from The Cancer Genome Atlas (TCGA) consortium as well as public data from several clinical trials in metastatic RCC were used for validation purposes. Multiregional sequencing data from the TRAcking Cancer Evolution through Therapy (TRACERx) RENAL cohort were used to assess the clonality of somatic mutations. Results: Of the 753 individuals with RCC identified in the MSK-IMPACT cohort, 115 showed evidence of targetable alterations, which represented a prevalence of 15.3% [95% confidence interval (CI), 12.7%–17.8%). When stratified by levels of evidence, the alterations identified corresponded to levels 2 (11.3%), 3A (5.2%), and 3B (83.5%). A low prevalence was recapitulated in the TCGA cohort at 9.1% (95% CI, 6.9%–11.2%). Copy-number variations predominated in papillary RCC tumors, largely due to amplifications in the MET gene. Notably, higher rates of actionability were found in individuals with metastatic disease (stage IV) compared with those with localized disease (OR, 2.50; 95% CI, 1.16–6.16; Fisher's P = 0.01). On the other hand, the prevalence of alterations associated with response to ICB therapy was found to be approximately 5% in both the MSK-IMPACT and TCGA cohorts and no associations with disease stage were identified (OR, 1.35; 95% CI, 0.46–5.40; P = 0.8). Finally, multiregional sequencing revealed that the vast majority of actionable mutations occurred later during tumor evolution and were only present subclonally in RCC tumors. Conclusions: RCC harbors a low prevalence of clinically actionable alterations compared with other tumors and the evidence supporting their clinical use is limited. These aberrations were found to be more common in advanced disease and seem to occur later during tumor evolution. Our study provides new insights on the role of targeted therapies for RCC and highlights the need for additional research to improve treatment selection using genomic profiling.
- Published
- 2020
123. Systemic therapy for advanced clear cell renal cell carcinoma after discontinuation of immune-oncology and VEGF targeted therapy combinations
- Author
-
Robert J. Motzer, Cihan Duzgol, Ritesh Kotecha, Andrea Knezevic, Brian I. Rini, Natalie Shapnik, Darren R. Feldman, Ruby Gupta, Martin H. Voss, Chung-Han Lee, Yasser Ged, Oguz Akin, and Sujata Patil
- Subjects
Oncology ,Sorafenib ,Adult ,Male ,medicine.medical_specialty ,Bevacizumab ,Survival ,Urology ,Salvage therapy ,Antineoplastic Agents ,lcsh:RC870-923 ,Pazopanib ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Internal medicine ,medicine ,Humans ,030212 general & internal medicine ,Carcinoma, Renal Cell ,IO combinations ,Aged ,Retrospective Studies ,Everolimus ,business.industry ,Sunitinib ,General Medicine ,Middle Aged ,lcsh:Diseases of the genitourinary system. Urology ,RCC ,VEGF ,Combined Modality Therapy ,Kidney Neoplasms ,Axitinib ,Receptors, Vascular Endothelial Growth Factor ,Reproductive Medicine ,chemistry ,Withholding Treatment ,030220 oncology & carcinogenesis ,Female ,Immunotherapy ,business ,Lenvatinib ,Immune-oncology ,medicine.drug ,Research Article - Abstract
Background Several phase 3 studies reported positive results for combinations of Immune-Oncology (IO) and Vascular Endothelial Growth Factor (VEGF) targeted therapies in patients with metastatic clear cell Renal Cell Carcinoma (ccRCC). However, there are limited data on outcomes to systemic therapy after IO-VEGF combinations. Methods A retrospective analysis was performed on patients with metastatic ccRCC treated at the Memorial Sloan Kettering Cancer Center and Cleveland Clinic who initiated systemic therapy post IO-VEGF including combinations with VEGF receptor (VEGFR) tyrosine kinase inhibitors (IO-TKI) and combinations with the anti-VEGF monoclonal antibody bevacizumab (IO-Bev). The study objectives were to evaluate the objective response rate (ORR), progression-free survival (PFS) and overall survival (OS) on systemic therapy post IO-VEGF. RECIST v1.1 criteria were used to determine radiological responses and progression. Survival estimates were evaluated with the Kaplan-Meier methods and the log-rank test from the start of systemic therapy post IO-VEGF to the event of interest. Results A total of fifty-nine patients were treated post discontinuation of IO-VEGF regimens which included IO-Bev (n = 35; 59%) and IO-TKI (n = 24; 41%). Fifty-eight patients (98%) received IO-VEGF regimens as part of a clinical trial. Subsequent therapies included cabozantinib (n = 22; 37%), axitinib (n = 18; 31%), pazopanib (n = 4; 7%), lenvatinib and everolimus (n = 4; 7%), mTOR inhibitor monotherapy (n = 3; 5%), axitinib and dalantercept (n = 2; 3%), sunitinib (n = 1; 2%), sorafenib (n = 1; 2%), and treatment with agents on unreported clinical trials (n = 4; 7%). Patients treated on unreported clinical trials were excluded from the efficacy analysis. Post IO-VEGF, the ORR was 25% and median PFS was 12.0 months (95% CI, 8.2–24.5). Median OS was 24.5 months (95% CI, 12–NE) and 12 months OS rate was 63.3% (95% CI, 48.6–74.9). We observed no differences post IO-VEGF OS when comparing IO- TKI vs IO-Bev (Log-rank p = 0.73). Conclusions Post IO-VEGF, most patients received VEGFR-TKIs. In this setting, VEGFR-TKIs demonstrated clinical activity and remain a viable option for salvage therapy after progression on IO-VEGF.
- Published
- 2020
124. PTEN Expression, Not Mutation Status in TSC1, TSC2, or mTOR, Correlates with the Outcome on Everolimus in Patients with Renal Cell Carcinoma Treated on the Randomized RECORD-3 Trial
- Author
-
Mahtab Marker, Jianning Xu, Almedina Redzematovic, Martin H. Voss, Jiayuan Shi, Ying-Bei Chen, David Chen, Parul Patel, Venkatraman E. Seshan, Irina Ostrovnaya, James J. Hsieh, Xia Han, A. Ari Hakimi, Albert Reising, and Robert J. Motzer
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Univariate analysis ,Everolimus ,biology ,business.industry ,Sunitinib ,medicine.disease ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Renal cell carcinoma ,030220 oncology & carcinogenesis ,Internal medicine ,Hepatocellular carcinoma ,Carcinoma ,biology.protein ,Medicine ,PTEN ,business ,PI3K/AKT/mTOR pathway ,medicine.drug - Abstract
Purpose: Genomic alterations in key components of PI3K/mTOR pathway have been proposed as candidate predictive markers for rapalog therapy in renal cell carcinoma (RCC). We tested this hypothesis in patients from a randomized phase II trial of everolimus versus sunitinib. Patients and Methods: Archival specimens collected at baseline were analyzed with targeted next-generation sequencing (NGS). Focus of interest were alterations in key PI3K pathway components. PTEN expression was assessed by IHC. Association between molecular findings and treatment outcomes was investigated; same associations were tested for 2 everolimus-treated trial cohorts in gastric and hepatocellular carcinoma (HCC). Results: Among 184 everolimus-treated patients with RCC with NGS data, mutation rates in genes of interest were 6% (TSC1), 4.4% (TSC2), and 8.2% (mTOR); 44% harbored alterations in ≥1 PI3K pathway component. For subjects with presence versus absence of mutations in TSC1, TSC2, or mTOR progression-free survival (PFS) neither differed on univariate analysis (HR, 1.0; P = 0.895) nor on multivariate testing stratified by MSKCC risk group and other established prognostic factors (HR, 1.1; P = 0.806). Everolimus-treated patients with retained (n = 50) versus lost (n = 50) PTEN IHC expression had median PFS of 5.3 months versus 10.5 months (HR, 2.5; P < 0.001). Such differences were not seen with sunitinib (10.9 months vs. 10.3 months; HR, 0.8; P = 0.475). Molecular findings did not correlate with outcomes in gastric and HCC cohorts. Conclusions: Association between mutation status for TSC1/TSC2/mTOR and therapeutic outcome on everolimus was not confirmed. Clinically meaningful differences in PFS were seen based on PTEN expression by IHC, lost in >50% of patients.
- Published
- 2019
- Full Text
- View/download PDF
125. Characterizing recurrent and lethal small renal masses in clear cell renal cell carcinoma using recurrent somatic mutations
- Author
-
Martin H. Voss, Ed Reznik, Daniel M. Tennenbaum, Brandon J. Manley, Jozefina Casuscelli, Mahyar Kashan, Paul Russo, Mazyar Ghanaat, Robert J. Motzer, Darren R. Feldman, Maria E. Arcila, A. Ari Hakimi, Maria I. Carlo, Jonathan A. Coleman, Almedina Redzematovic, Yusuke Sato, James J. Hsieh, and Maria F. Becerra
- Subjects
Male ,Oncology ,medicine.medical_specialty ,Urology ,Disease ,Article ,PBRM1 ,SETD2 ,Internal medicine ,Humans ,Medicine ,Carcinoma, Renal Cell ,Aged ,BAP1 ,business.industry ,Proportional hazards model ,Middle Aged ,medicine.disease ,Primary tumor ,Kidney Neoplasms ,Clear cell renal cell carcinoma ,Mutation ,Cohort ,Female ,business - Abstract
Introduction Small renal masses (SRMs) with evidence of clear cell renal cell carcinoma (ccRCC) are understudied. Current algorithms for the management of SRMs include surgical resection, ablation, and active surveillance. We sought to identify genomic biomarkers that could potentially refine the management of ccRCC in SRMs, especially in patients being evaluated for active surveillance. Methods We identified patients who had SRMs (4 cm or less) at time of surgery, had sequencing performed on their primary tumor and had a diagnosis of ccRCC. Patients were selected from 3 publicly available cohorts, The Cancer Genome Atlas ( n = 110), University of Tokyo ( n = 37), The International Cancer Genome Consortium ( n = 31), and from our own institutional prospective database ( n = 25). Among this cohort we analyzed mutations present in at least 5% of tumors, assessing for the enrichment of mutations and progression-free survival using the composite endpoint of recurrence or death of disease. Analysis was adjusted for multiple testing. A Cox regression model was used to assess clinical variables with significant mutations. Results In total, 203 patients were available for analysis. Median follow-up was 43.1 months among survivors. Mutations in VHL, PBRM1, SETD2, BAP1, KDM5C , and MTOR were present in more than 5% of tumors. Twenty-three patients (11.3%) had recurrence or died of their disease. Mutations in KDM5C were associated with inferior survival from either recurrence or death from disease, adjusted P 0.033. Conclusions We identified mutations in SRMs in ccRCC that are associated with recurrence and lethality. The strongest association was seen in those with KDM5C mutations. Use of these genomic biomarkers may improve stratification of patients with SRMs and for those who may be appropriate for active surveillance. Prospective evaluation of these markers is needed.
- Published
- 2019
- Full Text
- View/download PDF
126. PD39-05 PREVALENCE AND LANDSCAPE OF ACTIONABLE GENOMIC ALTERATIONS IN RENAL CELL CARCINOMA
- Author
-
Timothy A. Chan, Maria I. Carlo, Christopher J. Fong, Paul Russo, Nikolaus Schultz, Chung-Han Lee, Francisco Sanchez-Vega, Stanley Weng, A. Ari Hakimi, Renzo G. DiNatale, Eduard Reznik, Kyrollis Attalla, Martin H. Voss, Andrew W. Silagy, and Jonathan A. Coleman
- Subjects
Renal cell carcinoma ,business.industry ,Urology ,Cancer research ,medicine ,medicine.disease ,business - Abstract
INTRODUCTION AND OBJECTIVE:We report our experience with next-generation sequencing to characterize the prevalence and genomic landscape of actionable genomic alterations in renal cell carcinoma (R...
- Published
- 2020
- Full Text
- View/download PDF
127. MP14-15 THE SPINAL DISTRIBUTION OF METASTATIC RENAL CELL CARCINOMA: SUPPORT FOR LOCOREGIONAL RATHER THAN ARTERIAL HEMATOGENOUS MODE OF EARLY BONY DISSEMINATION
- Author
-
Martin H. Voss, Andrew W. Silagy, Kyrollis Attalla, Chung-Han Lee, Irina Ostrovnaya, Mark H. Bilsky, Nelson S Moss, Cihan Duzgol, Jonathan A. Coleman, Paul Russo, Renzo G. DiNatale, Lily McLaughlin, A. Ari Hakimi, Maria I. Carlo, and Jessica Flynn
- Subjects
Pathology ,medicine.medical_specialty ,Renal cell carcinoma ,business.industry ,Urology ,Mode (statistics) ,medicine ,Spinal metastasis ,Distribution (pharmacology) ,urologic and male genital diseases ,medicine.disease ,business - Abstract
INTRODUCTION AND OBJECTIVE:To investigate the distribution of spinal metastasis in metastatic renal cell carcinoma (mRCC) and to explore relationships between biological and clinical factors and pa...
- Published
- 2020
- Full Text
- View/download PDF
128. Phase 3 study of first-line treatment with pembrolizumab + belzutifan + lenvatinib or pembrolizumab/quavonlimab + lenvatinib versus pembrolizumab + lenvatinib for advanced renal cell carcinoma (RCC)
- Author
-
Toni K. Choueiri, Elizabeth R. Plimack, Thomas Powles, Martin H Voss, Howard Gurney, Rachel Kloss Silverman, Rodolfo F. Perini, Karla Rodriguez-Lopez, and Brian I. Rini
- Subjects
Cancer Research ,Oncology - Abstract
TPS399 Background: Combination therapy with the PD-1 inhibitor pembrolizumab and the vascular endothelial growth factor (VEGF) inhibitor lenvatinib showed antitumor activity as first-line treatment for advanced clear cell RCC (ccRCC) in the phase 3 KEYNOTE-581/CLEAR study (NCT02811861). Antitumor activity has also been shown with the hypoxia-inducible factor 2α (HIF-2α) inhibitor belzutifan (MK-6482) in ccRCC and with MK-1308A (coformulation of pembrolizumab and the CTLA-4 inhibitor quavonlimab) in non–small cell lung cancer. Therefore, HIF-2α or CTLA-4 inhibition with a PD-1 and VEGF inhibition backbone combination may provide additional benefit as first-line treatment in ccRCC. This open-label, randomized, phase 3 study (NCT04736706) will compare first-line treatment with the novel combination therapies pembrolizumab + belzutifan + lenvatinib (arm A) or MK-1308A + lenvatinib (arm B) with pembrolizumab + lenvatinib (arm C) for advanced RCC. Methods: Approximately 1,431 adults with metastatic ccRCC, measurable disease per RECIST v1.1, and KPS score ≥70% who had not previously undergone systemic therapy for advanced ccRCC will be enrolled. Patients will be randomly assigned 1:1:1 to arm A (belzutifan 120 mg + lenvatinib 20 mg orally once daily [QD] + pembrolizumab 400 mg IV every 6 weeks [Q6W]), arm B (MK-1308A [quavonlimab 25 mg + pembrolizumab 400 mg] IV [Q6W] and lenvatinib 20 mg orally QD), or arm C (pembrolizumab 400 mg IV [Q6W] + lenvatinib 20 mg orally QD). Treatment will continue until documented disease progression, withdrawal of consent, or other discontinuation event; patients will receive pembrolizumab and MK-1308A for up to 18 cycles (̃2 years). Stratification factors are International mRCC Database Consortium (IMDC) score (favorable vs intermediate vs poor), region of the world (North America vs Western Europe vs rest of world), and sarcomatoid features (yes vs no). Response will be assessed by CT or MRI per RECIST v1.1 by blinded independent central review (BICR) at week 12 from randomization, Q6W through week 78, and every 12 weeks thereafter. Adverse events and serious adverse events will be monitored throughout the study and for 90 days after treatment. Dual primary end points are progression-free survival per RECIST v1.1 by BICR and overall survival for arm A or arm B versus arm C in patients with IMDC intermediate/poor status and in all patients regardless of IMDC status. Secondary end points are objective response rate and duration of response per RECIST v1.1 by BICR, patient-reported outcomes, and safety. The study is recruiting patients at sites across, Asia, Australia, Europe, North America, and South America. Clinical trial information: NCT04736706.
- Published
- 2022
- Full Text
- View/download PDF
129. Part 1 results from NOVA-II, a randomized, double-blind, vehicle-controlled phase II study evaluating the safety and efficacy of OQL011 on VEGFR inhibitor associated hand-foot skin reaction in cancer patients
- Author
-
Mario E. Lacouture, Anisha B Patel, Nicole R. LeBoeuf, Omkar Subhash Marathe, Jonathan Leventhal, Jennifer N. Choi, Elaine Tat Lam, Benjamin S. Kaffenberger, Martin H Voss, Mark L Webb, Robert Claude Tyler, Juegang Ju, Hong Tang, Jie Luo, Milan J. Anadkat, and Nicholas J. Vogelzang
- Subjects
Cancer Research ,Oncology - Abstract
TPS396 Background: Hand-foot skin reaction (HFSR) is associated with the use of multi-targeted tyrosine kinase inhibitors sharing vascular endothelial growth factor receptor inhibitory (VEGFRi) activity (e.g., cabozantinib, regorafenib, sorafenib, sunitinib). HFSR affects the palms and soles with edema, erythema, hyperkeratosis, pain, and bullae/blisters impacting quality-of-life, activities of daily living, and consistent VEGFRi dosing. HFSR incidence differs among VEGFRi, ranging from 5-69% (all grades) and 1-19% (grade 3). Topical urea has marginal benefit, and to date, there are no regulatory authority approved HFSR treatments. The pathogenesis of HFSR has been partially associated with impaired vascular repair mechanisms resulting from inhibition of VEGF signaling. Nitric oxide (NO) has been reported to induce VEGF upregulation at a moderate concentration. We hypothesized that topical stimulation of VEGF signaling through OQL011, an NO donor, will safely and effectively mitigate HFSR. Methods: NOVA-II is a phase 2, double-blind, randomized vehicle-controlled trial evaluating the safety and efficacy of OQL011 for the treatment of moderate to severe HFSR. Subjects receiving a VEGFRi (either mono- or combination therapy) and have Grade ≥2 HFSR per NCI CTCAE v5.0 for palmar plantar erythrodysesthesia (PPE) were eligible. For Part 1, 31 subjects were randomized (2:1) and received 0.2% OQL011 topical ointment or vehicle TID for 6 weeks. The primary endpoint is the proportion of patients who achieve an NCI CTCAE v5.0 for PPE Grade 0 or 1 by Week 3. An investigator global assessment (IGA) scale is being developed to assess HFSR recovery, and a secondary endpoint will measure the proportion of patients who have improvement in HFSR severity and achieve an IGA score of clear (0) or almost clear (1) by Week 3 and/or Week 6. A Chi-square test will be used to detect a difference between the treatment arm and the vehicle-control arm. Additionally, the IGA score will be validated by assessing the inter- & intra-rater reliability. The correlation between IGA, NCI CTCAE v5.0 for PPE, and patient outcomes including Visual Analog Scale of Pain, HFSR Quality of Life questionnaire will also be reported. This study began enrolling patients in December 2019 and completed the last patient Visit in August 2021. Data lock, unblinding and data analysis will be completed by mid-November. Clinical trial information: NCT04088318.
- Published
- 2022
- Full Text
- View/download PDF
130. Comparative Genomic Profiling of Matched Primary and Metastatic Tumors in Renal Cell Carcinoma
- Author
-
Emily H. Cheng, Stephen B. Solomon, Darren R. Feldman, Ed Reznik, Renzo G. DiNatale, Philip Jonsson, Caitlin Bourque, Jonathan A. Coleman, Mazyar Ghanaat, Chung-Han Lee, Jozefina Casuscelli, Martin H. Voss, Jeremy C. Durack, Maria E. Arcila, Daniel M. Tennenbaum, A. Ari Hakimi, Brandon J. Manley, Robert J. Motzer, James J. Hsieh, Nick Socci, Maria I. Carlo, Almedina Redzematovic, Paul Russo, Kyle A. Blum, Mahyar Kashan, and Maria F. Becerra
- Subjects
Adult ,Male ,0301 basic medicine ,Oncology ,medicine.medical_specialty ,Pathology ,Lung Neoplasms ,Urology ,Concordance ,Adrenal Gland Neoplasms ,Bone Neoplasms ,Gene mutation ,medicine.disease_cause ,Article ,Germline ,Metastasis ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,Germline mutation ,Renal cell carcinoma ,Internal medicine ,medicine ,Humans ,Retroperitoneal Space ,Precision Medicine ,Carcinoma, Renal Cell ,Aged ,Histone Demethylases ,Mutation ,business.industry ,PTEN Phosphohydrolase ,High-Throughput Nucleotide Sequencing ,Genomics ,Histone-Lysine N-Methyltransferase ,Sequence Analysis, DNA ,Middle Aged ,medicine.disease ,Primary tumor ,Kidney Neoplasms ,030104 developmental biology ,Von Hippel-Lindau Tumor Suppressor Protein ,030220 oncology & carcinogenesis ,Female ,Lymph Nodes ,business - Abstract
Background Next-generation sequencing (NGS) studies of matched pairs of primary and metastatic tumors in renal cell carcinoma (RCC) have been limited to small cohorts. Objective To evaluate the discordance in somatic mutations between matched primary and metastatic RCC tumors. Design, setting, and participants Primary tumor (P), metastasis (M), and germline DNA from 60 patients with RCC was subjected to NGS with a targeted exon capture–based assay of 341 cancer-associated genes. Somatic mutations were called using a validated pipeline. Outcome measurements and statistical analysis Mutations were classified as shared (S) or private (Pr) in relation to each other within individual P-M pairs. The concordance score was calculated as (S−Pr)/(S+Pr). To calculate enrichment of Pr/S mutations for a particular gene, we calculated a two-sided p value from a binomial model for each gene with at least ten somatic mutation events, and also implemented a separate permutation test procedure. We adjusted p values for multiple hypothesis testing using the Benjamini-Hochberg procedure. The mutation discordance was calculated using Mann-Whitney U tests according to gene mutations or metastatic sites. Results and limitations Twenty-one pairs (35%) showed Pr mutations in both P and M samples. Of the remaining 39 pairs (65%), 14 (23%) had Pr mutations specific to P samples, 12 (20%) had Pr mutations to M samples, and 13 (22%) had identical somatic mutations. No individual gene mutation was preferentially enriched in either P or M samples. P-M pairs with SETD2 mutations demonstrated higher discordance than pairs with wild-type SETD2 . We observed that patients who received therapy before sampling of the P or M tissue had higher concordance of mutations for P-M pairs than patients who did not (Mann-Whitney p =0.088). Conclusions Our data show mutation discordance within matched P-M RCC tumor pairs. As most contemporary precision medicine trials do not differentiate mutations detected in P and M tumors, the prognostic and predictive value of mutations in P versus M tumors warrants further investigation. Patient summary In this study we evaluated the concordance of mutations between matched primary and metastatic tumors for 60 kidney cancer patients using a panel of 341 cancer genes. Forty-seven patients carried nonidentical cancer gene mutations within their matched primary-metastatic pair. The mutation profile of the primary tumor alone could compromise precision in selecting effective targeted therapies and result in suboptimal clinical outcomes.
- Published
- 2018
- Full Text
- View/download PDF
131. Durable response to anti-PD-1 immunotherapy in epithelioid angiomyolipoma: a report on the successful treatment of a rare malignancy
- Author
-
Martin H. Voss, Alisa N. Femia, Arjun Vasant Balar, Michael Lattanzi, Luis Chiriboga, Gopa Iyer, Shane A Meehan, Fang-Ming Deng, William C. Huang, and Yuliya Sundatova
- Subjects
Adult ,Male ,0301 basic medicine ,PD-L1 ,Cancer Research ,Angiomyolipoma ,medicine.medical_treatment ,Programmed Cell Death 1 Receptor ,Immunology ,Case Report ,Malignancy ,lcsh:RC254-282 ,03 medical and health sciences ,Tuberous sclerosis ,Antineoplastic Agents, Immunological ,0302 clinical medicine ,PD-1 ,medicine ,Humans ,Immunology and Allergy ,PEComa ,Everolimus ,Pharmacology ,biology ,business.industry ,Immunotherapy ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Kidney Neoplasms ,TSC2 ,Treatment Outcome ,030104 developmental biology ,Nivolumab ,Oncology ,030220 oncology & carcinogenesis ,mTOR ,biology.protein ,Cancer research ,Molecular Medicine ,business ,Epithelioid cell ,medicine.drug - Abstract
Background Malignant angiomyolipoma is an uncommon tumor of the class of perivasciular epithelioid cell neoplasms (PEComas). These tumors are characteristically driven by deleterious mutations in the tumor suppressors TSC1 and TSC2, whose gene products typically act to inhibit mTOR. There are several cases of malignant angiomyolipoma which exhibit transient responses to mTOR inhibitors, forming the basis of current practice guidelines in malignant PEComa. However the tumors ultimately acquire resistance, and there is no well-established second-line option. Despite the increasing prevalence of immunotherapy across a wide range of solid tumors, little is known about the immune infiltrate and PD-L1 expression of angiomyolipoma. Furthermore, there is no reported case on the treatment of malignant angiomyolipoma with an immune checkpoint inhibitor. Case presentation A 38 year-old man presented with gross hematuria and was diagnosed with renal epithelioid angiomyolipoma. Despite surgical resection, the tumor recurred and metastasized. Targeted genomic sequencing revealed a deleterious mutation in TSC2, and the patient was treated with the mTOR inihbitor everolimus. The patient went on to have a partial response but ultimately progressed. He was then treated with the anti-PD-1 immune checkpoint inhibitor nivolumab, and achieved a durable near-complete response which is ongoing after two years of treatment. Immunohistochemical staining of tumor tissue revealed strong PD-L1 expression and a brisk T-cell infiltrate. Conclusions We report on the first durable systemic treatment of malignant epithelioid angiomyolipoima with the use of PD-1 antibody nivolumab. Given the absence of prospective clinical trials in this exceedingly rare disease, particularly in the second-line setting, immune checkpoint inhibitors like nivolumab should be considered.
- Published
- 2018
- Full Text
- View/download PDF
132. Safety and efficacy of nivolumab in combination with sunitinib or pazopanib in advanced or metastatic renal cell carcinoma: the CheckMate 016 study
- Author
-
Lionel D. Lewis, David F. McDermott, Christian Kollmannsberger, Jennifer L. Spratlin, Marc S. Ernstoff, Michael A. Carducci, Brian I. Rini, Asim Amin, Daniel Y.C. Heng, Elmer Berghorn, Lingfeng Yang, Hans J. Hammers, Martin H. Voss, Elizabeth R. Plimack, Todd M. Bauer, and Jennifer J. Knox
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Immunology ,Metastatic renal cell carcinoma ,Tyrosine kinase inhibitor ,Ipilimumab ,Immune checkpoint inhibitor ,lcsh:RC254-282 ,Pazopanib ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine ,Sunitinib ,Immunology and Allergy ,Adverse effect ,Pharmacology ,business.industry ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Discontinuation ,Regimen ,030104 developmental biology ,Nivolumab ,Tolerability ,030220 oncology & carcinogenesis ,Antiangiogenic ,Molecular Medicine ,business ,medicine.drug ,Research Article - Abstract
Background Combination treatment with immune checkpoint inhibitors and antiangiogenic drugs has shown encouraging preliminary antitumor activity across various tumor types including advanced or metastatic renal cell carcinoma (aRCC). The open-label, parallel-cohort, dose-escalation, phase I CheckMate 016 study evaluated the efficacy and safety of nivolumab in combination with antiangiogenic tyrosine kinase inhibitors or ipilimumab. Long-term outcomes from this study for the combination of nivolumab plus sunitinib or pazopanib in aRCC are presented. Methods Patients with aRCC received nivolumab plus either sunitinib (50 mg/day, 4 weeks on/2 weeks off; N + S) or pazopanib (800 mg/day; N + P) until progression/unacceptable toxicity. The nivolumab starting dose was 2 mg/kg every 3 weeks, with planned escalation to 5 mg/kg every 3 weeks. Primary endpoints were safety and tolerability; antitumor activity was a secondary endpoint. Results Arm N + S enrolled 33 patients, 19 of whom were treatment-naïve; this arm advanced to the expansion phase. Median follow-up was 50.0 months. Patients experienced high frequencies of adverse events (AEs) including treatment-related AEs (100%), grade 3/4 treatment-related AEs (82%), and treatment-related AEs leading to discontinuation (39%). Investigator-assessed objective response rate (ORR) was 55% (18/33) and median progression-free survival (PFS) was 12.7 months. Median overall survival (OS) was not reached. Arm N + P enrolled 20 patients, all had ≥1 prior systemic therapy; this arm was closed due to dose-limiting toxicities and did not proceed to expansion. Median follow-up was 27.1 months. Patients treated with N + P experienced high frequencies of AEs including treatment-related AEs (100%), grade 3/4 treatment-related AEs (70%), and treatment-related AEs leading to discontinuation (25%). Investigator-assessed ORR was 45% (9/20) and median PFS was 7.2 months. Median OS was 27.9 months. Conclusions The addition of standard doses of sunitinib or pazopanib to nivolumab resulted in a high incidence of high-grade toxicities limiting future development of either combination regimen. While there was no adverse impact on response and the OS outcome was notable, the findings suggest that the success of combination regimens based on immune checkpoint inhibitors and antiangiogenic drugs may be dependent on careful selection of the antiangiogenic component and dose. Trial registration Clinicaltrials.gov identifier: NCT01472081. Registered 16 November 2011. Electronic supplementary material The online version of this article (10.1186/s40425-018-0420-0) contains supplementary material, which is available to authorized users.
- Published
- 2018
- Full Text
- View/download PDF
133. Genomic correlates of response to immune checkpoint therapies in clear cell renal cell carcinoma
- Author
-
William G. Kaelin, Dylan J. Martini, Charles G. Drake, Eliezer M. Van Allen, Marios Giannakis, Adam Tracy, Sabina Signoretti, Dana Cullen, Matthew D. Hellmann, Claire A. Margolis, Martin H. Voss, Mark W. Ball, Thai H. Ho, Diana Miao, Wei Li, Wenhua Gao, Dominick Bossé, Stephanie M. Wankowicz, Mohamad E. Allaf, Alexandra Snyder, Robert J. Motzer, F.S. Hodi, Megan Wind-Rotolo, Craig Norton, Christine Horak, and Toni K. Choueiri
- Subjects
0301 basic medicine ,Multidisciplinary ,business.industry ,medicine.medical_treatment ,Immunotherapy ,medicine.disease ,Immune checkpoint ,Chromatin remodeling ,PBRM1 ,03 medical and health sciences ,Clear cell renal cell carcinoma ,030104 developmental biology ,0302 clinical medicine ,030220 oncology & carcinogenesis ,medicine ,Cancer research ,Cytotoxic T cell ,business ,Transcription factor ,B7-H1 Antigen - Abstract
SNF'ing out antitumor immunity Immune checkpoint inhibitors induce durable tumor regressions in some, but not all, cancer patients. Understanding the mechanisms that determine tumor sensitivity to these drugs could potentially expand the number of patients who benefit (see the Perspective by Ghorani and Quezada). Pan et al. discovered that tumor cells in which a specific SWI/SNF chromatin remodeling complex had been experimentally inactivated were more sensitive to T cell–mediated killing. The cells were more responsive to interferon-γ, leading to increased secretion of cytokines that promote antitumor immunity. Miao et al. examined the genomic features of tumors from patients with metastatic renal cell carcinoma who had been treated with immune checkpoint inhibitors. Tumors harboring inactivating mutations in PBRM1 , which encodes a subunit of the same SWI/SNF complex, were more likely to respond to the drugs. Science , this issue p. 770 , p. 801 ; see also p. 745
- Published
- 2018
- Full Text
- View/download PDF
134. 417 Phase 3 study of pembrolizumab + belzutifan + lenvatinib or pembrolizumab/quavonlimab + lenvatinib versus pembrolizumab + lenvatinib as first-line treatment for advanced renal cell carcinoma
- Author
-
Elizabeth R. Plimack, Martin H. Voss, Brian I. Rini, Rachel Kloss Silverman, Rodolfo F. Perini, Thomas Powles, Howard Gurney, Karla Rodriguez-Lopez, and Toni K. Choueiri
- Subjects
Pharmacology ,Oncology ,Cancer Research ,medicine.medical_specialty ,Randomization ,business.industry ,Immunology ,Phases of clinical research ,Pembrolizumab ,medicine.disease ,Discontinuation ,chemistry.chemical_compound ,Clear cell renal cell carcinoma ,chemistry ,Renal cell carcinoma ,Internal medicine ,medicine ,Molecular Medicine ,Immunology and Allergy ,Lenvatinib ,business ,Adverse effect - Abstract
BackgroundPembrolizumab + vascular endothelial growth factor (VEGF) inhibitor lenvatinib demonstrated antitumor activity as first-line treatment for advanced clear cell renal cell carcinoma (ccRCC) in phase 3 trial KEYNOTE-581/CLEAR (NCT02811861). Hypoxia-inducible factor 2α (HIF-2α) inhibitor belzutifan (MK-6482) showed antitumor activity in ccRCC, and a coformulation of pembrolizumab and CTLA-4 inhibitor quavonlimab (MK-1308A) showed antitumor activity in non–small cell lung cancer. HIF-2α or CTLA-4 inhibition with PD-1 and VEGF inhibition backbone combination may provide additional benefit as first-line treatment in ccRCC. This open-label, randomized, phase 3 study (NCT04736706) will be conducted to compare novel combination therapies pembrolizumab + belzutifan + lenvatinib (arm A) and MK-1308A + lenvatinib (arm B) with pembrolizumab + lenvatinib (arm C).MethodsApproximately 1431 adults with metastatic ccRCC, measurable disease per RECIST v1.1, and Karnofsky Performance Status Scale score ≥70% who had not previously undergone systemic therapy for advanced ccRCC will be enrolled. Patients will be randomly assigned 1:1:1 to arm A (belzutifan 120 mg + lenvatinib 20 mg oral once daily + pembrolizumab 400 mg IV every 6 weeks), arm B (MK-1308A [quavonlimab 25 mg + pembrolizumab 400 mg] IV every 6 weeks and lenvatinib 20 mg oral once daily), or arm C (pembrolizumab 400 mg IV every 6 weeks + lenvatinib 20 mg oral once daily). Treatment will continue until documented disease progression, withdrawal of consent, or other discontinuation event; patients will receive pembrolizumab and MK-1308A for up to 18 cycles (approximately 2 years). Patients will be stratified by International mRCC Database Consortium (IMDC) score (favorable vs intermediate vs poor), region of the world (North America vs Western Europe vs rest of the world), and sarcomatoid features (yes vs no). Response will be assessed by CT or MRI per RECIST v1.1 by blinded independent central review (BICR) at week 12 from randomization, every 6 weeks through week 78, and every 12 weeks thereafter. Adverse events and serious adverse events will be monitored throughout the study and for 90 days after treatment. Dual primary end points are progression-free survival per RECIST v1.1 by BICR and overall survival. Primary end points will be assessed in arm A compared with arm C and in arm B compared with arm C for patients with IMDC intermediate/poor status and in all patients regardless of IMDC status. Secondary end points are objective response rate and duration of response per RECIST v1.1 by BICR, patient-reported outcomes, and safety.AcknowledgementsMedical writing and/or editorial assistance was provided by Matthew Grzywacz, PhD, of ApotheCom (Yardley, PA, USA). This assistance was funded by Merck Sharp & Dohme Corp., a subsidiary of Merck & Co., Inc., Kenilworth, NJ, USA, and Eisai Inc., Woodcliff Lake, NJ, USA. Funding for this research was provided by Merck Sharp & Dohme Corp., a subsidiary of Merck & Co., Inc., Kenilworth, NJ, USA,Eisai Inc., Woodcliff Lake, NJ, USA.Trial RegistrationClinicaltrials.gov, NCT04736706Ethics ApprovalThe study and the protocol were approved by the Institutional Review Board or ethics committee at each site.
- Published
- 2021
- Full Text
- View/download PDF
135. Clinical and Morphologic Characteristics of Fibroblast Growth Factor Receptor Inhibitor–Associated Retinopathy
- Author
-
Murk Heinemann, Gopa Iyer, Alison M. Schram, Ghassan K. Abou-Alfa, Jonathan E. Rosenberg, Martin H. Voss, James J. Harding, Julia Canestraro, Alexander Drilon, Anuja Kriplani, Komal Jhaveri, David H. Abramson, Dianna Haggag-Lindgren, Dean F. Bajorin, and Jasmine H. Francis
- Subjects
medicine.medical_specialty ,Visual acuity ,genetic structures ,Visual Acuity ,Angiogenesis Inhibitors ,chemistry.chemical_compound ,Pharmacotherapy ,Retinal Diseases ,Interquartile range ,Ophthalmology ,medicine ,Humans ,Adverse effect ,Protein Kinase Inhibitors ,Aged ,Retrospective Studies ,Aged, 80 and over ,business.industry ,Brief Report ,Subretinal Fluid ,Retinal ,Middle Aged ,medicine.disease ,Receptors, Fibroblast Growth Factor ,eye diseases ,Serous fluid ,chemistry ,Fibroblast growth factor receptor ,Female ,sense organs ,medicine.symptom ,business ,Tomography, Optical Coherence ,Retinopathy - Abstract
Importance Fibroblast growth factor receptor (FGFR) 1 to 4 inhibitors are approved by the US Food and Drug Administration and suppress the mitogen-activated protein kinase (MAPK) pathway, with a potential for treatment-related retinopathy. To date, implications of FGFR inhibitor–associated ocular toxic effects are poorly described. Therefore, more detailed clinical descriptions of this ocular toxic effect could help explain visual symptoms while receiving drug therapy. Objective To describe the clinical and morphologic characteristics of serous retinal disturbances associated with FGFR inhibitors. Design, Setting, and Participants In this retrospective case series, 146 patients receiving FGFR inhibitors as cancer treatment at a single tertiary referral center were included. This study included 40 eyes of 20 patients with retinopathy by optical coherence tomography (OCT). OCTs were obtained on the remaining patients and the results were judged normal. Patients were recruited from March 2012 to January 2021. Main Outcomes and Measures Characteristics of treatment-emergent choroidal and retinal OCT abnormalities as compared with baseline OCT, associated with visual acuity at presentation and at fluid resolution. Results A total of 20 of 146 patients (13.7%) exhibited FGFR inhibitor–associated retinopathy. Of these 20 patients, 11 (55%) were female, and the median (range) age was 62.6 (42.7-86.0) years. The median (range; mean) time from medication start to initial subretinal fluid detection was 21 (5-125; 32) days. The median (interquartile range [IQR]) baseline logMAR best-corrected visual acuity (BCVA) was 0 (0-0.1). At fluid accumulation, 11 eyes had decreased vision: the median (IQR) subgroup baseline BCVA was 0 (0-0.1); and the median (IQR) BCVA change from baseline to accumulation was −0.1 (−0.2 to −0.1). For 26 eyes (65%) with follow-up, the subretinal fluid resolved without medical intervention or drug interruption in all but 1 patient. At fluid resolution, the median (IQR) BCVA was 0.1 (0-0.1), and the change in median (IQR) BCVA from baseline to fluid resolution was 0 (−0.03 to 0). No patient discontinued drug therapy on account of their retinopathy. Conclusions and Relevance FGFR inhibitors result in subretinal fluid foci similar to other drugs that inhibit the MAPK pathway. In this series, FGFR inhibitors did not cause irreversible loss of vision; the retinopathy was self-limited and did not require medical intervention. These results may explain visual symptoms while taking the drug, although the precise frequency or magnitude of this adverse effect cannot be determined with certainty from this retrospective investigation.
- Published
- 2021
- Full Text
- View/download PDF
136. Tumor Xenografts of Human Clear Cell Renal Cell Carcinoma But Not Corresponding Cell Lines Recapitulate Clinical Response to Sunitinib: Feasibility of Using Biopsy Samples
- Author
-
Song Han, Nicole Benfante, Martin H. Voss, Chung-Han Lee, Jeremy C. Durack, Maria F. Becerra, Maria E. Arcila, John H. Healey, James J. Hsieh, Mohit Chawla, Brandon J. Manley, Nicola Fabbri, Yiyu Dong, A. Ari Hakimi, Omer Aras, Robert J. Motzer, Patrick J. Boland, Almedina Redzematovic, Jozefina Casuscelli, Jonathan A. Coleman, Ying-Bei Chen, Ed Reznik, Daniel M. Tennenbaum, Emily H. Cheng, Darren R. Feldman, and Paul Russo
- Subjects
Male ,0301 basic medicine ,medicine.medical_specialty ,Pathology ,Urology ,medicine.medical_treatment ,Transplantation, Heterologous ,Antineoplastic Agents ,Mice, SCID ,Kidney ,Article ,Targeted therapy ,Random Allocation ,03 medical and health sciences ,0302 clinical medicine ,Mice, Inbred NOD ,Renal cell carcinoma ,Cell Line, Tumor ,Biopsy ,Sunitinib ,medicine ,Animals ,Humans ,Carcinoma, Renal Cell ,Aged ,medicine.diagnostic_test ,business.industry ,Biopsy, Needle ,Histology ,Middle Aged ,medicine.disease ,Xenograft Model Antitumor Assays ,Kidney Neoplasms ,Clear cell renal cell carcinoma ,Treatment Outcome ,030104 developmental biology ,030220 oncology & carcinogenesis ,Cancer research ,Feasibility Studies ,Heterografts ,Female ,Histopathology ,business ,Clear cell ,medicine.drug - Abstract
Background Parallel development of preclinical models that recapitulate treatment response observed in patients is central to the advancement of personalized medicine. Objective To evaluate the use of biopsy specimens to develop patient-derived xenografts and the use of corresponding cell lines from renal cell carcinoma (RCC) tumors for the assessment of histopathology, genomics, and treatment response. Design, setting, and participants A total of 74 tumor specimens from 66 patients with RCC were implanted into immunocompromised NOD- SCID IL2Rg −/− mice. Four cell lines generated from patients' specimens with clear cell pathology were used for comparative studies. Outcome measurements and statistical analysis Preclinical models were established and assessed. Engraftment rates were analyzed using chi-square testing. Analysis of variance (two-way analysis of variance) was conducted to assess tumor growth. Results and limitations Overall, 33 RCC mouse xenograft models were generated with an overall engraftment rate of 45% (33 of 74). Tumor biopsies engrafted comparably with surgically resected tumors (58% vs 41%; p =0.3). Xenograft tumors and their original tumors showed high fidelity in regard to histology, mutation status, copy number change, and targeted therapy response. Engraftment rates from metastatic tumors were higher but not more significant than primary tumors (54% vs 34%; p =0.091). Our engraftment rate using metastases or biopsies was comparable with recent reports using resected primary tumors. In stark contrast to corresponding cell lines, all tested xenografts recapitulated patients' clinical response to sunitinib. Conclusions Patient-derived xenograft models can be effectively established from tumor biopsies. Preclinical xenograft models but not matched cell lines reflected clinical responses to sunitinib. Patient summary Matched patient-derived clear cell renal cell carcinoma xenografts and cell lines from responsive and refractory patients treated with sunitinib were established and evaluated for pharmacologic response to anti–vascular endothelial growth factor treatment. Both models accurately reflected the genetic characteristics of original tumors, but only xenografts recapitulated drug responses observed in patients. These models could serve as a powerful platform for precision medicine.
- Published
- 2017
- Full Text
- View/download PDF
137. Genomic Characterization of Renal Medullary Carcinoma and Treatment Outcomes
- Author
-
Darren R. Feldman, Alison M. Schram, Yelena Kemel, Ying-Bei Chen, Martin H. Voss, A. Ari Hakimi, Joshua Chaim, Kaitlin M. Woo, Maria I. Carlo, Robert J. Motzer, James J. Hsieh, Gouri Nanjangud, Sujata Patil, Devyn Taylor Coskey, and Chung-Han Lee
- Subjects
Adult ,Male ,0301 basic medicine ,Oncology ,medicine.medical_specialty ,Pathology ,Adolescent ,Pharmacogenomic Variants ,Urology ,Translocation, Genetic ,Article ,Renal medullary carcinoma ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine ,Humans ,SMARCB1 ,Child ,In Situ Hybridization, Fluorescence ,Survival analysis ,Aged ,Platinum ,Retrospective Studies ,Sickle cell trait ,medicine.diagnostic_test ,business.industry ,Retrospective cohort study ,SMARCB1 Protein ,Sequence Analysis, DNA ,Middle Aged ,medicine.disease ,Immunohistochemistry ,Survival Analysis ,Kidney Neoplasms ,Treatment Outcome ,030104 developmental biology ,Medullary carcinoma ,Carcinoma, Medullary ,030220 oncology & carcinogenesis ,Female ,business ,Kidney cancer ,Fluorescence in situ hybridization - Abstract
Background Renal medullary carcinoma (RMC) is a rare and aggressive type of kidney cancer that primarily affects young adults with sickle cell trait; outcomes are poor despite treatment. Identifying molecular features of this tumor could provide biologic rationale for novel targeted therapies. The objective was to report on clinical outcomes with systemic therapy and characterize molecular features. Patients and Methods This was a retrospective analysis on 36 patients given a pathologic diagnosis of RMC at one institution from 1995 to 2015. Tumors were analyzed for expression of SWI/SNF Related, Matrix Associated, Actin Dependent Regulator Of Chromatin, Subfamily B, Member 1 (SMARCB1) through immunohistochemistry and for genomic alterations with fluorescence in situ hybridization for SMARCB1, and targeted next-generation sequencing. Time from initiation of therapy to progression of disease and overall survival were calculated using the Kaplan–Meier method. Results The median age in the cohort was 28 (range, 12-72) years, and all patients tested had sickle cell trait. Overall survival was 5.8 months (95% confidence interval [CI], 4.1-10.9) and for 12 patients who received platinum-based therapy, median progression-free survival was 2.5 months (95% CI, 1.2-not reached). A total of 10 available tumors underwent analysis with fluorescence in situ hybridization for SMARCB1; this revealed loss of heterozygosity with concurrent translocation in 8, and biallelic loss in 2. Next-generation targeted sequencing showed no recurring mutations. Conclusions Outcome was generally poor in this cohort of patients with RMC. Uniform loss of SMARCB1 is a key molecular feature in this tumor and mechanism of loss appears to be mostly through translocations and deletions.
- Published
- 2017
- Full Text
- View/download PDF
138. OncoKB: A Precision Oncology Knowledge Base
- Author
-
José Baselga, Feras M. Hantash, Tiffany A. Traina, Antonio M. P. Omuro, James J. Harding, Julia E. Rudolph, Margaret K. Callahan, Daniel C. Danila, Rona Yaeger, Alan L. Ho, Ederlinda Paraiso, Hongxin Zhang, Michael F. Berger, Ritika Kundra, Ahmet Zehir, Leonard B. Saltz, Ping Chi, Douglas A. Levine, Gregory J. Riely, Neerav Shukla, David M. Hyman, Moriah H. Nissan, Alexandra Snyder, Mrinal Gounder, Yelena Y. Janjigian, Maeve A. Lowery, Matthew D. Hellmann, Debyani Chakravarty, Tara Soumerai, Sarah Fierberg Phillips, Marc Ladanyi, Shrujal S. Baxi, Andrew Grupe, Thomas Kaley, Barry S. Taylor, Jiaojiao Wang, Martin H. Voss, Paul Sabbatini, David B. Solit, Dana Rathkopf, Alexander N. Shoushtari, Nikolaus Schultz, Gopa Iyer, Jianjiong Gao, Paul K. Paik, Michael A. Postow, Sarat Chandarlapaty, and Matthew T. Chang
- Subjects
0301 basic medicine ,Cancer Research ,business.industry ,Specific mutation ,MEDLINE ,Cancer ,Evidence-based medicine ,Bioinformatics ,medicine.disease ,Expert group ,Article ,Food and drug administration ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Oncology ,Knowledge base ,Precision oncology ,030220 oncology & carcinogenesis ,Medicine ,business - Abstract
Purpose With prospective clinical sequencing of tumors emerging as a mainstay in cancer care, an urgent need exists for a clinical support tool that distills the clinical implications associated with specific mutation events into a standardized and easily interpretable format. To this end, we developed OncoKB, an expert-guided precision oncology knowledge base. Methods OncoKB annotates the biologic and oncogenic effects and prognostic and predictive significance of somatic molecular alterations. Potential treatment implications are stratified by the level of evidence that a specific molecular alteration is predictive of drug response on the basis of US Food and Drug Administration labeling, National Comprehensive Cancer Network guidelines, disease-focused expert group recommendations, and scientific literature. Results To date, > 3,000 unique mutations, fusions, and copy number alterations in 418 cancer-associated genes have been annotated. To test the utility of OncoKB, we annotated all genomic events in 5,983 primary tumor samples in 19 cancer types. Forty-one percent of samples harbored at least one potentially actionable alteration, of which 7.5% were predictive of clinical benefit from a standard treatment. OncoKB annotations are available through a public Web resource ( http://oncokb.org ) and are incorporated into the cBioPortal for Cancer Genomics to facilitate the interpretation of genomic alterations by physicians and researchers. Conclusion OncoKB, a comprehensive and curated precision oncology knowledge base, offers oncologists detailed, evidence-based information about individual somatic mutations and structural alterations present in patient tumors with the goal of supporting optimal treatment decisions.
- Published
- 2017
- Full Text
- View/download PDF
139. Integration of Recurrent Somatic Mutations with Clinical Outcomes: A Pooled Analysis of 1049 Patients with Clear Cell Renal Cell Carcinoma
- Author
-
Yusuke Sato, Darren R. Feldman, Jonathan A. Coleman, Paul Russo, Robert J. Motzer, Masashi Fukayama, Haruki Kume, Martin H. Voss, Daniel M. Tennenbaum, Brandon J. Manley, Nicole Benfante, Maria F. Becerra, Teppei Morikawa, Emily C. Zabor, James J. Hsieh, Maria E. Arcila, Seishi Ogawa, A. Ari Hakimi, Victor E. Reuter, Almedina Redzematovic, Jozefina Casuscelli, and Yukio Homma
- Subjects
Adult ,Male ,0301 basic medicine ,Oncology ,medicine.medical_specialty ,Urology ,Gene mutation ,Bioinformatics ,Logistic regression ,medicine.disease_cause ,03 medical and health sciences ,0302 clinical medicine ,Risk Factors ,Renal cell carcinoma ,Internal medicine ,Biomarkers, Tumor ,medicine ,Humans ,Prospective Studies ,Prospective cohort study ,Carcinoma, Renal Cell ,Aged ,Neoplasm Staging ,Aged, 80 and over ,Mutation ,BAP1 ,business.industry ,Tumor Suppressor Proteins ,Confounding ,Histone-Lysine N-Methyltransferase ,Sequence Analysis, DNA ,Middle Aged ,Prognosis ,medicine.disease ,Kidney Neoplasms ,Clear cell renal cell carcinoma ,030104 developmental biology ,030220 oncology & carcinogenesis ,Female ,Neoplasm Recurrence, Local ,business ,Ubiquitin Thiolesterase - Abstract
Background Analyses of associations between clinicopathologic outcomes and recurrent somatic mutations in clear cell renal cell carcinoma (ccRCC) have been limited to individual cohorts. Objective To define clinicopathologic associations between specific mutations and ccRCC disease characteristics. Design, setting, and participants DNA sequencing data were pooled from three collaborative genomic cohorts ( n =754) and our institutional database ( n =295). All patients had clinical data and identification of somatic mutations from their primary tumors. Outcome measurements and statistical analysis Analysis of gene mutations for associations with maximal tumor size (linear regression) and pathologic stage (logistic regression). Cancer-specific survival (CSS) and recurrence-free survival (RFS) were calculated using competing risks methods. Analyses were adjusted for cohort site, and results were adjusted for multiple testing ( q value). Relevant genes were used in multivariable models that included confounding variables and the validated Mayo Clinic Stage, Size, Grade, and Necrosis (SSIGN) score. Results and limitations Association with tumor size was found for mutations in BAP1 ( q =0.013). No mutations were found to be associated with stage after adjusted analysis. Mutations in BAP1 ( q =0.004) and TP53 ( q =0.001) were associated with decreased CSS in a multivariable model; only TP53 ( q =0.005) remained significant when SSIGN score was included. SETD2 mutations ( q =0.047) were associated with decreased RFS in multivariable models, including models with SSIGN score. Conclusions In >1000 patients with ccRCC, pooled analysis and multivariable modeling demonstrated that three mutated genes have statistically significant associations with poor clinical outcomes. This included the more commonly mutated BAP1 and SETD2 and the less frequently mutated TP53 . After adjustment for clinical confounders, mutations of TP53 and SETD2 were associated with decreased CSS and RFS, respectively. Patient summary Using rigorous statistical methods, this study affirmed that certain mutations in clear cell renal cell carcinoma may portend inferior survival and an increased risk of recurrence.
- Published
- 2017
- Full Text
- View/download PDF
140. The PARP Inhibitor Veliparib Can Be Safely Added to Bendamustine and Rituximab and Has Preliminary Evidence of Activity in B-Cell Lymphoma
- Author
-
Jacob D. Soumerai, Andrew D. Zelenetz, Craig H. Moskowitz, M. Lia Palomba, Paul A. Hamlin, Ariela Noy, David J. Straus, Alison J. Moskowitz, Anas Younes, Matthew J. Matasar, Steven M. Horwitz, Carol S. Portlock, Jason A. Konner, Mrinal M. Gounder, David M. Hyman, Martin H. Voss, Matthew G. Fury, Devika Gajria, Richard D. Carvajal, Alan L. Ho, Jan H. Beumer, Brian Kiesel, Zhigang Zhang, Alice Chen, Richard F. Little, Christine Jarjies, Thu O. Dang, Fallon France, Nishant Mishra, and John F. Gerecitano
- Subjects
Adult ,Male ,0301 basic medicine ,Oncology ,Bendamustine ,Cancer Research ,medicine.medical_specialty ,Lymphoma, B-Cell ,Maximum Tolerated Dose ,Veliparib ,Poly(ADP-ribose) Polymerase Inhibitors ,Neutropenia ,Disease-Free Survival ,Drug Administration Schedule ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,hemic and lymphatic diseases ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Bendamustine Hydrochloride ,Humans ,B-cell lymphoma ,Multiple myeloma ,Aged ,Leukopenia ,Dose-Response Relationship, Drug ,business.industry ,medicine.disease ,Surgery ,Lymphoma ,030104 developmental biology ,chemistry ,030220 oncology & carcinogenesis ,Benzimidazoles ,Female ,Rituximab ,medicine.symptom ,business ,medicine.drug - Abstract
Purpose: The PARP inhibitor veliparib enhances the cytotoxicity of alkylating agents. This phase I study evaluated veliparib with the bifunctional alkylator bendamustine (VB) in patients with relapsed/refractory lymphoma, multiple myeloma, and solid malignancies, with a cohort expansion of VB with rituximab (VBR) in patients with B-cell lymphomas. Experimental Design: This dose-escalation study evaluated safety, pharmacokinetics, and preliminary efficacy of veliparib (20–400 mg twice a day, days 1–7 of 28-day cycle) and bendamustine (70 and 90 mg/m2 intravenously, days 1 and 2). A cohort expansion was conducted, which combined veliparib and bendamustine at the maximum tolerated dose (MTD) with rituximab (375 mg/m2, day 1) in patients with B-cell lymphomas. Thirty-four patients were treated in seven dose-escalation cohorts and seven patients in the dose-expansion cohort. Results: The MTD was veliparib 300 mg twice daily plus bendamustine 90 mg/m2. Dose-limiting toxicities (DLT) were anemia, nausea, hypertension, and hyperhidrosis. Grade ≥3 toxicities included lymphopenia (87.8%), anemia (19.5%), neutropenia (12.2%), thrombocytopenia (9.8%), leukopenia (9.8%), nausea (7.3%), and hypophosphatemia (7.3%). Apparent veliparib clearance was slightly lower than previously reported. Of 14 patients with lymphoma evaluable for response, five of seven (71%) on VB and six of seven (86%) on VBR achieved objective response. One patient with multiple myeloma achieved partial response. Conclusions: VB and VBR were generally well-tolerated. VBR had preliminary clinical activity in patients with B-cell lymphoma, which warrants further investigation in a phase II trial. This trial was registered at www.clinicaltrials.gov as NCT01326702. Clin Cancer Res; 23(15); 4119–26. ©2017 AACR.
- Published
- 2017
- Full Text
- View/download PDF
141. Genomic Alterations and Outcomes with VEGF-Targeted Therapy in Patients with Clear Cell Renal Cell Carcinoma
- Author
-
Chung-Han Lee, Martin H. Voss, A.A. Hakimi, Kaitlin M. Woo, Sujata Patil, Robert J. Motzer, Ying-Bei Chen, William Lee, Darren R. Feldman, M. Ladanyi, James J. Hsieh, Maria I. Carlo, Almedina Redzematovic, Devyn Taylor Coskey, Maria E. Arcila, and Brandon J. Manley
- Subjects
0301 basic medicine ,Oncology ,Research Report ,Clear cell renal cell carcinoma ,medicine.medical_specialty ,medicine.medical_treatment ,Disease ,medicine.disease_cause ,Systemic therapy ,PBRM1 ,Targeted therapy ,03 medical and health sciences ,0302 clinical medicine ,SETD2 ,Internal medicine ,medicine ,genomics ,Mutation ,BAP1 ,business.industry ,medicine.disease ,3. Good health ,VEGF-targeted therapy ,030104 developmental biology ,Nephrology ,030220 oncology & carcinogenesis ,prognosis ,business - Abstract
Background: Mutations in VHL, PBRM1, SETD2, BAP1, and KDM5C are common in clear cell renal cell carcinoma (ccRCC), and presence of certain mutations has been associated with outcomes in patients with non-metastatic disease. Limited information is available regarding the correlation between genomic alterations and outcomes in patients with metastatic disease, including response to VEGF-targeted therapy. Objective: To explore correlations between mutational profiles and cancer-specific outcomes, including response to standard VEGF-targeted agents, in patients with metastatic cc RCC. Methods: A retrospective review of 105 patients with metastatic ccRCC who had received systemic therapy and had targeted next-generation sequencing of tumors was conducted. Genomic alterations were correlated to outcomes, including overall survival and time to treatment failure to VEGF-targeted therapy. Results: The most frequent mutations were detected in VHL (83%), PBRM1 (51%), SETD2 (35%), BAP1 (24%), KDM5C (16%), and TERT (14%). Time to treatment failure with VEGF-targeted therapy differed significantly by PBRM1 mutation status (p = 0.01, median 12.0 months for MT versus 6.9 months for WT) and BAP1 mutation status (p = 0.01, median 6.4 months for MT versus 11.0 months for WT). Shorter overall survival was associated with TERT mutations (p = 0.03, median 29.6 months for MT versus 52.6 months for WT) or BAP1 mutations (p = 0.02, median 28.7 months for MT versus not reached for WT). Conclusions: Genomic alterations in ccRCC tumors have prognostic implications in patients with metastatic disease. BAP1 and TERT promoter mutations may be present in higher frequency than previously thought, and based on this data, deserve further study for their association with poor prognosis.
- Published
- 2017
142. Genomic Biomarkers of a Randomized Trial Comparing First-line Everolimus and Sunitinib in Patients with Metastatic Renal Cell Carcinoma
- Author
-
Helen Won, S. Duygu Selcuklu, A. Ari Hakimi, David Chen, Michael F. Berger, Patrizia Pinciroli, Agnes Viale, Nancy Bouvier, Patricia Wang, Ying-Bei Chen, Umesh Bhanot, Parul Patel, Michael Chevinsky, William Lee, Nicholas D. Socci, Jennifer J. Knox, Almedina Redzematovic, Maurizio Voi, Emily H. Cheng, Nils Weinhold, Mahtab Marker, Robert J. Motzer, James J. Hsieh, Martin H. Voss, Daoqi You, and Kety Huberman
- Subjects
Male ,0301 basic medicine ,Oncology ,Pathology ,Indoles ,medicine.medical_treatment ,Targeted therapy ,0302 clinical medicine ,Renal cell carcinoma ,Sunitinib ,Randomized Controlled Trials as Topic ,Aged, 80 and over ,Histone Demethylases ,BAP1 ,Nuclear Proteins ,Middle Aged ,Prognosis ,Kidney Neoplasms ,DNA-Binding Proteins ,Survival Rate ,Von Hippel-Lindau Tumor Suppressor Protein ,030220 oncology & carcinogenesis ,Female ,Ubiquitin Thiolesterase ,medicine.drug ,Adult ,medicine.medical_specialty ,Urology ,Antineoplastic Agents ,Disease-Free Survival ,Young Adult ,03 medical and health sciences ,Internal medicine ,medicine ,Humans ,Pyrroles ,Everolimus ,Progression-free survival ,Carcinoma, Renal Cell ,Aged ,Proportional Hazards Models ,business.industry ,Tumor Suppressor Proteins ,PTEN Phosphohydrolase ,Histone-Lysine N-Methyltransferase ,medicine.disease ,Clear cell renal cell carcinoma ,030104 developmental biology ,Mutation ,business ,Kidney cancer ,Transcription Factors - Abstract
Background Metastatic renal cell carcinoma (RCC) patients are commonly treated with vascular endothelial growth factor (VEGF) inhibitors or mammalian target of rapamycin inhibitors. Correlations between somatic mutations and first-line targeted therapy outcomes have not been reported on a randomized trial. Objective To evaluate the relationship between tumor mutations and treatment outcomes in RECORD-3, a randomized trial comparing first-line everolimus (mTOR inhibitor) followed by sunitinib (VEGF inhibitor) at progression with the opposite sequence in 471 metastatic RCC patients. Design, setting, and participants Targeted sequencing of 341 cancer genes at ∼540× coverage was performed on available tumor samples from 258 patients; 220 with clear cell histology (ccRCC). Outcome measurements and statistical analysis Associations between somatic mutations and median first-line progression free survival (PFS1L) and overall survival were determined in metastatic ccRCC using Cox proportional hazards models and log-rank tests. Results and limitations Prevalent mutations (≥ 10%) were VHL (75%), PBRM1 (46%), SETD2 (30%), BAP1 (19%), KDM5C (15%), and PTEN (12%). With first-line everolimus, PBRM1 and BAP1 mutations were associated with longer (median [95% confidence interval {CI}] 12.8 [8.1, 18.4] vs 5.5 [3.1, 8.4] mo) and shorter (median [95% CI] 4.9 [2.9, 8.1] vs 10.5 [7.3, 12.9] mo) PFS1L, respectively. With first-line sunitinib, KDM5C mutations were associated with longer PFS1L (median [95% CI] of 20.6 [12.4, 27.3] vs 8.3 [7.8, 11.0] mo). Molecular subgroups of metastatic ccRCC based on PBRM1 , BAP1 , and KDM5C mutations could have predictive values for patients treated with VEGF or mTOR inhibitors. Most tumor DNA was obtained from primary nephrectomy samples (94%), which could impact correlation statistics. Conclusions PBRM1 , BAP1 , and KDM5C mutations impact outcomes of targeted therapies in metastatic ccRCC patients. Patient summary Large-scale genomic kidney cancer studies reported novel mutations and heterogeneous features among individual tumors, which could contribute to varied clinical outcomes. We demonstrated correlations between somatic mutations and treatment outcomes in clear cell renal cell carcinoma, supporting the value of genomic classification in prospective studies.
- Published
- 2017
- Full Text
- View/download PDF
143. A pan-cancer analysis of PBAF complex mutations and their association with immunotherapy response
- Author
-
A Ari, Hakimi, Kyrollis, Attalla, Renzo G, DiNatale, Irina, Ostrovnaya, Jessica, Flynn, Kyle A, Blum, Yasser, Ged, Douglas, Hoen, Sviatoslav M, Kendall, Ed, Reznik, Anita, Bowman, Jason, Hwee, Christopher J, Fong, Fengshen, Kuo, Martin H, Voss, Timothy A, Chan, and Robert J, Motzer
- Subjects
Adult ,Aged, 80 and over ,Chromosomal Proteins, Non-Histone ,Kaplan-Meier Estimate ,Middle Aged ,Kidney Neoplasms ,Cohort Studies ,DNA-Binding Proteins ,Gene Expression Regulation, Neoplastic ,Mutation ,Humans ,Immunotherapy ,Carcinoma, Renal Cell ,Aged ,Transcription Factors - Abstract
There is conflicting data regarding the role of PBAF complex mutations and response to immune checkpoint blockade (ICB) therapy in clear cell renal cell carcinoma (ccRCC) and other solid tumors. We assess the prevalence of PBAF complex mutations from two large cohorts including the pan-cancer TCGA project (n = 10,359) and the MSK-IMPACT pan-cancer immunotherapy cohort (n = 3700). Across both cohorts, PBAF complex mutations, predominantly PBRM1 mutations, are most common in ccRCC. In multivariate models of ccRCC patients treated with ICB (n = 189), loss-of-function (LOF) mutations in PBRM1 are not associated with overall survival (OS) (HR = 1.24, p = 0.47) or time to treatment failure (HR = 0.85, p = 0.44). In a series of 11 solid tumors (n = 2936), LOF mutations are not associated with improved OS in a stratified multivariate model (HR = 0.9, p = 0.7). In a current series of solid tumors treated with ICB, we are unable to demonstrate favorable response to ICB in patients with PBAF complex mutations.
- Published
- 2019
144. The society for immunotherapy of cancer consensus statement on immunotherapy for the treatment of advanced renal cell carcinoma (RCC)
- Author
-
David I. Quinn, Virginia Seery, Christopher G. Wood, Robert J. Motzer, Allan J. Pantuck, Laura S. Wood, Hans J. Hammers, Michael B. Atkins, Martin H. Voss, Daniel J. George, Thomas E. Hutson, Dena Battle, Brian I. Rini, Sumanta K. Pal, Eric Jonasch, Richard W. Joseph, David F. McDermott, and Robert A. Figlin
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,medicine.medical_treatment ,Immunology ,Ipilimumab ,Pembrolizumab ,Guidelines ,lcsh:RC254-282 ,Avelumab ,03 medical and health sciences ,0302 clinical medicine ,Antineoplastic Agents, Immunological ,Cancer immunotherapy ,Internal medicine ,Position Article and Guidelines ,medicine ,Biomarkers, Tumor ,Immunology and Allergy ,Humans ,Molecular Targeted Therapy ,Immune checkpoint inhibitor (ICI) ,Carcinoma, Renal Cell ,Neoplasm Staging ,Pharmacology ,business.industry ,Renal cell carcinoma (RCC) ,Disease Management ,Kidney cancer ,Immunotherapy ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,Kidney Neoplasms ,Axitinib ,030104 developmental biology ,030220 oncology & carcinogenesis ,Practice Guidelines as Topic ,Molecular Medicine ,Nivolumab ,business ,medicine.drug - Abstract
The approval of immunotherapeutic agents and immunotherapy-based combination strategies in recent years has revolutionized the treatment of patients with advanced renal cell carcinoma (aRCC). Nivolumab, a programmed death 1 (PD-1) immune checkpoint inhibitor monoclonal antibody, was approved as monotherapy in 2015 for aRCC after treatment with a VEGF-targeting agent. In April 2018, the combination of nivolumab and ipilimumab, a CTLA-4 inhibitor, was approved for intermediate- and poor-risk, previously untreated patients with aRCC. Then, in 2019, combinations therapies consisting of pembrolizumab (anti-PD-1) or avelumab (anti-PD-ligand (L) 1) with axitinib (a VEGF receptor tyrosine kinase inhibitor) were also approved to treat aRCC and are likely to produce dramatic shifts in the therapeutic landscape. To address the rapid advances in immunotherapy options for patients with aRCC, the Society for Immunotherapy of Cancer (SITC) reconvened its Cancer Immunotherapy Guidelines (CIG) Renal Cell Carcinoma Subcommittee and tasked it with generating updated consensus recommendations for the treatment of patients with this disease.
- Published
- 2019
145. Systemic therapy for metastatic renal cell carcinoma-is timing everything?
- Author
-
Martin H. Voss and Ritesh Kotecha
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,business.industry ,Treatment outcome ,Cancer ,General Medicine ,Disease ,medicine.disease ,Medical care ,Systemic therapy ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Renal cell carcinoma ,030220 oncology & carcinogenesis ,Internal medicine ,Localized disease ,Curative surgery ,Medicine ,business - Abstract
Timeliness of medical care for patients with newly diagnosed cancer has long been recognized as a cornerstone metric for healthcare quality. For patients with renal cell carcinoma (RCC), such considerations have largely focused on patients with localized disease awaiting curative surgery (1,2). Delays, though, often occur for patients with metastatic disease, particularly in those with symptomatic disease, and may also affect treatment outcomes.
- Published
- 2019
146. Results from a randomised phase I/II trial evaluating the safety and antitumour activity of anti-PD-1 (MEDI0680)/anti-PD-L1 (durvalumab) vs anti-PD-1 (nivolumab) alone in metastatic clear cell renal cell carcinoma (ccRCC)
- Author
-
L. Lewis, S.J. Welsh, H. Hu, Sjoukje F. Oosting, Ikbel Achour, Aaron R. Hansen, F.L. Walcott, A.A. Azad, Martin H. Voss, Jhanelle E. Gray, and Guided Treatment in Optimal Selected Cancer Patients (GUTS)
- Subjects
medicine.medical_specialty ,Durvalumab ,business.industry ,Disease progression ,Anti pd 1 ,Hematology ,EPIC ,Phase i ii ,Oncology ,Family medicine ,medicine ,Disease characteristics ,Nivolumab ,business ,Measurable Lesion - Abstract
Background MEDI0680 is a humanised IgG4κ anti-programmed cell death-1 (PD-1) mAb. We hypothesised that simultaneous blockade of PD-1:PD-L1/PD-L2 with MEDI0680 (M) and PD-1:PD-L1/CD80 with anti-PD-L1 mAb durvalumab (D) would improve efficacy vs blockade of the PD-1:PD-L1/PD-L2 pathway (with nivolumab; N) alone. M+D was well tolerated in the dose-escalation phase of a Phase I/II study in pts with advanced solid tumours, with an ORR of 33% (10/30; including 3/4 RCC pts). In the Phase II portion of the study, we compared M+D to N in a dose-expansion cohort of pretreated, immunotherapy (IO)-naive pts with metastatic ccRCC. Methods Eligible pts had received 1–3 prior therapy lines, no prior IO exposure and ≥1 measurable lesion. They were randomised 2:1 (stratified by MSKCC risk group and PD-L1 expression) to M 20 mg/kg IV + D 750 mg Q2W or N 240 mg IV Q2W until unacceptable toxicity or disease progression, for ≤2 years. Endpoints included investigator-assessed ORR by RECIST v1.1 (primary endpoint), PFS and safety (secondary). Sample size was ∼60 to detect a difference of 26.0% (ie, ORR = 47.5%, assuming ORR of 21.5% for N) with 76% power at a 1-sided significance level of 0.10. Results By Feb 24, 2019, 63 pts were randomised. Baseline pt/disease characteristics were generally well balanced, but more pts on N had favourable MSKCC risk (7/21; 33.3%) vs M+D (10/42; 23.8%). ORR was 14.3% (6/42; 2 CR, 4 PR; plus 2 unconfirmed PR) vs 19.0% (4/21; 4 PR, 0 unconfirmed) for M+D and N, respectively. There was no difference between arms in ORR by PD-L1 expression ( Conclusions Efficacy was similar with combined M+D and N monotherapy in pts with TKI-pretreated, IO-naive, metastatic ccRCC, but more pts discontinued M+D due to TRAEs. Clinical trial identification NCT02118337. Editorial acknowledgement Aaron Korpal, PhD, of Cirrus Communications (Macclesfield, UK), an Ashfield company, funded by AstraZeneca. Legal entity responsible for the study AstraZeneca. Funding AstraZeneca. Disclosure M.H. Voss: Research grant / Funding (institution): BMS; Research grant / Funding (institution): Genentech; Honoraria (self): Eisai; Honoraria (self): Exelixis; Honoraria (self): Pfizer; Honoraria (self): Novartis; Honoraria (self): Calithera; Honoraria (self): Corvus. A.A. Azad: Honoraria (self), Advisory / Consultancy: Janssen; Honoraria (self), Advisory / Consultancy, Research grant / Funding (institution), Non-remunerated activity/ies: Astellas; Honoraria (self), Advisory / Consultancy: Novartis; Research grant / Funding (institution), Non-remunerated activity/ies: Merck Serono; Honoraria (self), Advisory / Consultancy: Tolmar; Honoraria (self), Advisory / Consultancy, Non-remunerated activity/ies: Amgen; Honoraria (self), Advisory / Consultancy: Pfizer; Honoraria (self): Bayer; Honoraria (self), Advisory / Consultancy: Telix Pharmaceuticals; Honoraria (self), Advisory / Consultancy: Bristol-Myers Squibb; Honoraria (self), Advisory / Consultancy: Sanofi. A.R. Hansen: Advisory / Consultancy, Research grant / Funding (institution): Genentech/Roche; Advisory / Consultancy, Research grant / Funding (institution): Merck; Advisory / Consultancy, Research grant / Funding (institution): GSK; Advisory / Consultancy, Research grant / Funding (institution): Bristol-Myers Squibb; Advisory / Consultancy, Research grant / Funding (institution): Novartis; Research grant / Funding (institution): Boston Biomedical; Advisory / Consultancy, Research grant / Funding (institution): Boehringer Ingelheim; Advisory / Consultancy, Research grant / Funding (institution): AstraZeneca. J.E. Gray: Honoraria (self), Advisory / Consultancy, Research grant / Funding (institution): AstraZeneca; Honoraria (self), Advisory / Consultancy, Research grant / Funding (institution): Genentech; Honoraria (self), Advisory / Consultancy: Celgene; Honoraria (self), Advisory / Consultancy: Takeda; Honoraria (self), Advisory / Consultancy: Janssen; Honoraria (self), Advisory / Consultancy: Eli Lilly; Honoraria (self), Advisory / Consultancy: Triptych Health Partners; Research grant / Funding (institution): Array; Research grant / Funding (institution): Merck; Research grant / Funding (institution): Epic Sciences; Research grant / Funding (institution): BMS; Research grant / Funding (institution): BI; Research grant / Funding (institution): Trovagene; Research grant / Funding (institution): Loxo; Research grant / Funding (institution): Blueprint; Research grant / Funding (institution): Novartis. I. Achour: Shareholder / Stockholder / Stock options, Full / Part-time employment: AstraZeneca. H. Hu: Full / Part-time employment: AstraZeneca. L. Lewis: Travel / Accommodation / Expenses, Full / Part-time employment: AstraZeneca. F.L. Walcott: Shareholder / Stockholder / Stock options, Full / Part-time employment: AstraZeneca. S.F. Oosting: Research grant / Funding (institution): Celldex; Research grant / Funding (institution): Novartis. All other authors have declared no conflicts of interest.
- Published
- 2019
147. Putative Drivers of Aggressiveness in TCEB1-mutant Renal Cell Carcinoma: An Emerging Entity with Variable Clinical Course
- Author
-
Roy Mano, Victor E. Reuter, Alex Sanchez, Vladimir Makarov, Robert J. Motzer, A. Ari Hakimi, Benjamin A Freeman, Kyrollis Attalla, Alexander N. Gorelick, Jonathan A. Coleman, Andrew W. Silagy, Ed Reznik, Renzo G. DiNatale, Martin H. Voss, Timothy A. Chan, Stanley Weng, Diego Chowell, Ying-Bei Chen, Satish K. Tickoo, Paul Russo, Julian Marcon, and Kyle A. Blum
- Subjects
Oncology ,medicine.medical_specialty ,DNA Copy Number Variations ,Somatic cell ,Urology ,030232 urology & nephrology ,Article ,Metastasis ,03 medical and health sciences ,0302 clinical medicine ,Germline mutation ,Interquartile range ,Renal cell carcinoma ,Internal medicine ,medicine ,Biomarkers, Tumor ,Humans ,Carcinoma, Renal Cell ,business.industry ,Genomics ,Sequence Analysis, DNA ,medicine.disease ,Phenotype ,Kidney Neoplasms ,030220 oncology & carcinogenesis ,Cohort ,business ,Kidney cancer - Abstract
Background TCEB1-mutant renal cell carcinoma (RCC) is a rare variant of RCC with clear-cell features. Owing to its unique morphological and molecular features it has recently been proposed as a separate entity. Initial series suggested an indolent, early-stage phenotype. Here we expand our clinical cohort and describe a more detailed genomic analysis looking for potential drivers of aggressiveness. Design, setting, and participants We identified five new cases in our institutional sequencing cohort, four of whom were found to have high-stage disease (American Joint Committee on Cancer stage III/IV). Twelve previously reported cases were pooled for comparison purposes (Sato, The Cancer Genome Atlas, TRACERx Renal). Outcome measures and statistical analysis We used our previously validated pipeline to analyze somatic mutations and copy number alterations (CNAs) in seven tumor samples with available data and estimated the number of cancer cells bearing each somatic mutation. The oncogenic potential of mutations was assessed using OncoKB and two other algorithms. Mann-Whitney U tests were used to evaluate differences in genomic markers between stage groups. Results and limitations All tumors showed biallelic inactivation of the TCEB1 gene according to a combination of somatic mutation and CNA analyses. Mutations were always found in residues involved in hydrophobic interactions with VHL. We found that high-stage tumors had additional oncogenic mutations (median 1, interquartile range [IQR] 1–1 vs 2, IQR 2–2; median difference 1, 95% confidence interval [CI] 1–1; p = 0.002) and showed whole-genome doubling events. They also seemed to have a higher burden of somatic CNAs (median fraction CNA genome 0.10, IQR 0.10–0.15 vs 0.63, IQR 0.58–0.68), however, this finding did not reach statistical significance (median difference 0.49, 95% CI 0.33–0.63; p = 0.052). Conclusions TCEB1-mutant RCC can show variable behavior ranging from very indolent to aggressive. Specific molecular events leading to high genomic instability seem to be associated with aggressiveness. This study expands the clinical spectrum of TCEB1-mutant RCC. Patient summary We present four cases of aggressive TCEB1-mutant renal cell carcinoma, a rare type of kidney cancer. In-depth analysis of the genomes of these tumors revealed certain abnormalities that might explain this aggressive behavior.
- Published
- 2019
148. First-in-Humans Trial of Dasatinib-Derivative Tracer for Tumor Kinase-Targeted PET
- Author
-
Eva Burnazi, Daniel C. Danila, Bradley J. Beattie, Serge K. Lyashchenko, Darren R. Veach, Wolfgang A. Weber, Pat Zanzonico, Michael Mauro, Lukas M. Carter, Martin H. Voss, Mark Dunphy, Monica Fornier, Christina Pressl, Steven M. Larson, Simone Krebs, Milan Grkovski, Kevin Staton, Jason S. Lewis, and Manda Null
- Subjects
0301 basic medicine ,Male ,Biodistribution ,Fluorine Radioisotopes ,Dasatinib ,Pilot Projects ,Effective dose (radiation) ,03 medical and health sciences ,0302 clinical medicine ,In vivo ,Neoplasms ,Positron Emission Tomography Computed Tomography ,Medicine ,Dosimetry ,Humans ,Radiology, Nuclear Medicine and imaging ,Whole Body Imaging ,Prospective Studies ,Aged ,Kidney ,business.industry ,Middle Aged ,Protein-Tyrosine Kinases ,medicine.disease ,Leukemia ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Absorbed dose ,Female ,Radiopharmaceuticals ,business ,Nuclear medicine ,medicine.drug - Abstract
We developed a first-of-kind dasatinib-derivative imaging agent, (18)F-SKI-249380 ((1)(8)F-SKI), and validated its use for noninvasive in vivo tyrosine kinase-targeted tumor detection in preclinical models. In this study, we assessed the feasibility of using (18)F-SKI for PET imaging in patients with malignancies. Methods: Five patients with a prior diagnosis of breast cancer, renal cell cancer, or leukemia underwent whole-body PET/CT imaging 90 min after injection of (18)F-SKI (mean, 241.24 ± 116.36 MBq) as part of a prospective study. In addition, patients underwent either a 30-min dynamic scan of the upper abdomen including, at least partly, cardiac left ventricle, liver, spleen, and kidney (n = 2) or three 10-min whole-body PET/CT scans (n = 3) immediately after injection and blood-based radioactivity measurements to determine the time course of tracer distribution and facilitate radiation dose estimates. A subset of 3 patients had a delayed whole-body PET/CT scan at 180 min. Biodistribution, dosimetry, and tumor uptake were quantified. Absorbed doses were calculated using OLINDA/EXM 1.0. Results: No adverse events occurred after injection of (18)F-SKI. In total, 27 tumor lesions were analyzed, with a median SUV(peak) of 1.4 (range, 0.7–2.3) and tumor-to-blood ratios of 1.6 (range, 0.8–2.5) at 90 min after injection. The intratumoral drug concentrations calculated for 4 reference lesions ranged from 0.03 to 0.07 nM. In all reference lesions, constant tracer accumulation was observed between 30 and 90 min after injection. A blood radioassay indicated that radiotracer clearance from blood and plasma was initially rapid (blood half-time, 1.31 ± 0.81 min; plasma, 1.07 ± 0.66 min; n = 4), followed variably by either a prolonged terminal phase (blood half-time, 285 ± 148.49 min; plasma, 240 ± 84.85 min; n = 2) or a small rise to a plateau (n = 2). Like dasatinib, (18)F-SKI underwent extensive metabolism after administration, as evidenced by metabolite analysis. Radioactivity was predominantly cleared via the hepatobiliary route. The highest absorbed dose estimates (mGy/MBq) in normal tissues were to the right colon (0.167 ± 0.04) and small intestine (0.153 ± 0.03). The effective dose was 0.0258 mSv/MBq (SD, 0.0034 mSv/MBq). Conclusion: (18)F-SKI demonstrated significant tumor uptake, distinct image contrast despite low injected doses, and rapid clearance from blood.
- Published
- 2019
149. Towards individualized therapy for metastatic renal cell carcinoma
- Author
-
Robert J. Motzer, Martin H. Voss, and Ritesh Kotecha
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,business.industry ,Disease ,medicine.disease ,Kidney Neoplasms ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Renal cell carcinoma ,030220 oncology & carcinogenesis ,Internal medicine ,medicine ,Carcinoma ,Humans ,Molecular Targeted Therapy ,Precision Medicine ,business ,Carcinoma, Renal Cell ,Biomarkers - Abstract
Over the past decade, the treatment landscape for patients with metastatic renal cell carcinoma (RCC) has evolved dramatically. The therapeutic options available have expanded and now include immune-checkpoint inhibitors, novel targeted agents and combination strategies, and thus optimal patient selection and treatment sequencing are increasingly pertinent for optimizing clinical outcomes. A better understanding of the underlying biology of the tumour and its microenvironment continues to drive the inception of new diagnostic and therapeutic approaches. Furthermore, many biomarkers robustly associated with treatment and disease-specific outcomes have been identified, and their integration into clinical decision-making for patients with advanced-stage disease will soon become a reality. Herein, we review relevant aspects of the molecular biology of metastatic RCC, with an emphasis on predictive and prognostic biomarkers, and suggest tailored algorithms to individualize and guide treatment approaches for specific subgroups of patients.
- Published
- 2019
150. A phase 2, randomized trial evaluating the combination of dalantercept plus axitinib in advanced clear cell renal cell carcinoma
- Author
-
Robert S. Alter, J. Thaddeus Beck, Monika Joshi, Earle F. Burgess, David R. Shaffer, Mayer Fishman, Ralph J. Hauke, Nicholas J. Vogelzang, Martin H. Voss, Nauman Moazzam, Xiaosha Zhang, Chad E. Glasser, Elizabeth R. Plimack, Rupal S. Bhatt, Michael B. Atkins, Theodore F. Logan, Rahul A. Parikh, Matthew L. Sherman, and Brian I. Rini
- Subjects
Adult ,Diarrhea ,Male ,Cancer Research ,medicine.medical_specialty ,Randomization ,Axitinib ,Activin Receptors, Type II ,Recombinant Fusion Proteins ,Urology ,Angiogenesis Inhibitors ,Placebo ,Article ,law.invention ,03 medical and health sciences ,0302 clinical medicine ,Randomized controlled trial ,Double-Blind Method ,Renal cell carcinoma ,law ,Antineoplastic Combined Chemotherapy Protocols ,Clinical endpoint ,Medicine ,Humans ,030212 general & internal medicine ,Carcinoma, Renal Cell ,Fatigue ,Aged ,Aged, 80 and over ,business.industry ,Hazard ratio ,Middle Aged ,medicine.disease ,Kidney Neoplasms ,Progression-Free Survival ,Immunoglobulin Fc Fragments ,Clear cell renal cell carcinoma ,Oncology ,030220 oncology & carcinogenesis ,Hypertension ,Female ,business ,medicine.drug - Abstract
BACKGROUND In a prior open-label study, the combination of dalantercept, a novel antiangiogenic targeting activin receptor-like kinase 1 (ALK1), plus axitinib was deemed safe and tolerable with a promising efficacy signal in patients with advanced renal cell carcinoma (RCC). METHODS In the current phase 2, randomized, double-blind, placebo-controlled study, patients with clear cell RCC previously treated with 1 prior angiogenesis inhibitor were randomized 1:1 to receive axitinib plus dalantercept versus axitinib plus placebo. Randomization was stratified by the type of prior therapy. The primary endpoint was progression-free survival (PFS). Secondary endpoints were PFS in patients with ≥2 prior lines of anticancer therapy, overall survival, and the objective response rate. RESULTS Between June 10, 2014, and February 23, 2017, a total of 124 patients were randomly assigned to receive axitinib plus dalantercept (59 patients) or placebo (65 patients). The median PFS was not found to be significantly different between the treatment groups (median, 6.8 months vs 5.6 months; hazard ratio, 1.11 [95% CI, 0.71-1.73; P = .670]). Neither group reached the median overall survival (hazard ratio, 1.39 [95% CI, 0.70-2.77; P = .349]). The objective response rate was 19.0% (11 of 58 patients; 95% CI, 9.9%-31.4%) in the dalantercept plus axitinib group and 24.6% (15 of 61 patients; 95% CI, 14.5%-37.3%) in the placebo plus axitinib group. At least 1 treatment-emergent adverse event of ≥grade 3 was observed in 59% of patients (34 of 58 patients) in the dalantercept group and 64% of patients (39 of 61 patients) in the placebo group. One treatment-related death occurred in the placebo plus axitinib group. CONCLUSIONS Although well tolerated, the addition of dalantercept to axitinib did not appear to improve treatment-related outcomes in previously treated patients with advanced RCC.
- Published
- 2019
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.