3151
|
Hart S, Gupta G. Editorial on "Biochemical and Epigenetic Insights into L-2-Hydroxyglutarate, a Potential Therapeutic Target in Renal Cancer". ANNALS OF TRANSLATIONAL MEDICINE 2019; 6:S105. [PMID: 30740426 DOI: 10.21037/atm.2018.11.43] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Spencer Hart
- Department of Urology, Loyola University Medical Center, Maywood, IL, USA
| | - Gopal Gupta
- Department of Urology, Loyola University Medical Center, Maywood, IL, USA
| |
Collapse
|
3152
|
Recent success and limitations of immune checkpoint inhibitors for cancer: a lesson from melanoma. Virchows Arch 2019; 474:421-432. [PMID: 30747264 DOI: 10.1007/s00428-019-02538-4] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 01/20/2019] [Accepted: 02/01/2019] [Indexed: 02/08/2023]
Abstract
Several researches have been carried over the last few decades to understand of how cancer evades the immune system and thus to identify therapies that could directly act on patient's immune system in the way of restore or induce a response to cancer. As a consequence, "cancer immunotherapy" is conquering predominantly the modern scenario of the fight against cancer. The recent clinical success of immune checkpoint inhibitors (ICIs) has created an entire new class of anti-cancer drugs and restored interest in the field of immuno-oncology, leading to regulatory approvals of several agents for the treatment of a variety of malignancies. The first to be approved in 2011 was the anti-CTLA-4 antibody ipilimumab for the treatment of unresectable or metastatic melanoma. Subsequently, the anti-PD-1s, nivolumab and pembrolizumab, received regulatory approvals for the treatment of melanoma and several other cancers. More recently, three anti-PD-L1 antibodies have received approval: atezolizumab and durvalumab for locally advanced or metastatic urothelial carcinoma and metastatic non-small cell lung cancer (NSCLC) and avelumab for the treatment of locally advanced or metastatic urothelial carcinoma and metastatic Merkel cell carcinoma. This review, starting from the results of melanoma trials, highlights in turn different ICIs and data for different indications in several malignancies are included under each drug class.
Collapse
|
3153
|
Haque W, Aghazadeh M, Miles BJ, Satkunasivam R, Butler EB, Teh BS. Clinical benefit of treatment for metastatic renal cell cancer at high volume facilities. ANNALS OF TRANSLATIONAL MEDICINE 2019; 6:S90. [PMID: 30740411 DOI: 10.21037/atm.2018.11.02] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Waqar Haque
- Department of Radiation Oncology, Houston Methodist Hospital, Houston, TX, USA
| | - Monty Aghazadeh
- Department of Urology, Houston Methodist Hospital, Houston, TX, USA
| | - Brian J Miles
- Department of Urology, Houston Methodist Hospital, Houston, TX, USA
| | - Raj Satkunasivam
- Department of Urology, Houston Methodist Hospital, Houston, TX, USA
| | - E Brian Butler
- Department of Radiation Oncology, Houston Methodist Hospital, Houston, TX, USA
| | - Bin S Teh
- Department of Radiation Oncology, Houston Methodist Hospital, Houston, TX, USA
| |
Collapse
|
3154
|
Mutational and Antigenic Landscape in Tumor Progression and Cancer Immunotherapy. Trends Cell Biol 2019; 29:396-416. [PMID: 30765144 DOI: 10.1016/j.tcb.2019.01.003] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 01/04/2019] [Accepted: 01/08/2019] [Indexed: 12/18/2022]
Abstract
Evolving neoplasms accumulate non-synonymous mutations at a high rate, potentially enabling the expression of antigenic epitopes that can be recognized by the immune system. Since they are not covered by central tolerance, such tumor neoantigens (TNAs) should be under robust immune control as they surge. However, genetic defects that impair cancer cell eradication by the immune system coupled with the establishment of local immunosuppression can enable TNA accumulation, which is generally associated with improved clinical sensitivity to various immunotherapies. Here, we explore how tumor-intrinsic factors and immunological processes shape the mutational and antigenic landscape of evolving neoplasms to influence clinical responses to immunotherapy, and propose strategies to achieve robust immunological control of the disease despite disabled immunosurveillance.
Collapse
|
3155
|
Kong BY, Bolton H, Kim JW, Silveira PA, Fromm PD, Clark GJ. On the Other Side: Manipulating the Immune Checkpoint Landscape of Dendritic Cells to Enhance Cancer Immunotherapy. Front Oncol 2019; 9:50. [PMID: 30788290 PMCID: PMC6372550 DOI: 10.3389/fonc.2019.00050] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 01/17/2019] [Indexed: 12/26/2022] Open
Abstract
Monoclonal antibodies targeting co-inhibitory immune checkpoint molecules have been successful in clinical trials of both solid and hematological malignancies as acknowledged by the 2018 Nobel Prize in Medicine, however improving clinical response rates is now key to expanding their efficacy in areas of unmet medical need. Antibodies to checkpoint inhibitors target molecules on either T cells or tumor cells to stimulate T cells or remove tumor mediated immunosuppression, respectively. However, many of the well-characterized T cell immune checkpoint receptors have their ligands on antigen presenting cells or exert direct effects on those cells. Dendritic cells are the most powerful antigen presenting cells; they possess the ability to elicit antigen-specific responses and have important roles in regulation of immune tolerance. Despite their theoretical benefits in cancer immunotherapy, the translation of DC therapies into the clinic is yet to be fully realized and combining DC-based immunotherapy with immune checkpoint inhibitors is an attractive strategy. This combination takes advantage of the antigen presenting capability of DC to maximize specific immune responses to tumor antigens whilst removing tumor-associated immune inhibitory mechanisms with immune checkpoint inhibition. Here we review the expression and functional effects of immune checkpoint molecules on DC and identify rational combinations for DC vaccination to enhance antigen-specific T cell responses, cytokine production, and promotion of long-lasting immunological memory.
Collapse
Affiliation(s)
- Benjamin Y Kong
- Dendritic Cell Research Group, ANZAC Research Institute, Concord, NSW, Australia.,Sydney Medical School, The University of Sydney, Camperdown, NSW, Australia.,Department of Medical Oncology, Concord Repatriation General Hospital, Concord, NSW, Australia
| | - Holly Bolton
- Dendritic Cell Research Group, ANZAC Research Institute, Concord, NSW, Australia.,Sydney Medical School, The University of Sydney, Camperdown, NSW, Australia
| | - Julius W Kim
- Dendritic Cell Research Group, ANZAC Research Institute, Concord, NSW, Australia.,Sydney Medical School, The University of Sydney, Camperdown, NSW, Australia
| | - Pablo A Silveira
- Dendritic Cell Research Group, ANZAC Research Institute, Concord, NSW, Australia.,Sydney Medical School, The University of Sydney, Camperdown, NSW, Australia
| | - Phillip D Fromm
- Dendritic Cell Research Group, ANZAC Research Institute, Concord, NSW, Australia.,Sydney Medical School, The University of Sydney, Camperdown, NSW, Australia
| | - Georgina J Clark
- Dendritic Cell Research Group, ANZAC Research Institute, Concord, NSW, Australia.,Sydney Medical School, The University of Sydney, Camperdown, NSW, Australia
| |
Collapse
|
3156
|
Canino C, Perrone L, Bosco E, Saltalamacchia G, Mosca A, Rizzo M, Porta C. Targeting angiogenesis in metastatic renal cell carcinoma. Expert Rev Anticancer Ther 2019; 19:245-257. [PMID: 30678509 DOI: 10.1080/14737140.2019.1574574] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
INTRODUCTION Renal cell carcinoma (RCC), and particularly its clear cell histological subtype, is commonly characterized by genetic alterations in the Von Hippel Lindau (VHL) tumor suppressor gene, leading to a typically exasperated angiogenesis. However, other biological and genetic peculiarities contribute to differentiate this malignancy from other solid tumors, including its immunogenicity. Areas covered: This review focuses on the present and future role of antiangiogenic drugs, administered either alone (as it has been in the past few years), or in combination with other agents (e.g. immune checkpoint inhibitors), in the treatment of metastatic RCC. Expert commentary: Due to its peculiar pathogenesis, it is unrealistic to expect to be able to get rid of antiangiogenic agents for the treatment of this disease; however, we do expect that combinations of VEGF/VEGFRs-targeting agents with immune checkpoint inhibitors will gradually replace antiangiogenic monotherapies as the standard of care, at least in the first line setting of metastatic RCC patients. Biomarkers discovery remains the highest priority in order to further improve the percentage of patients benefitting of our treatment.
Collapse
Affiliation(s)
- Costanza Canino
- a Division of Translational Oncology , I.R.C.C.S. Istituti Clinici Scientifici Maugeri , Pavia , Italy
| | - Lorenzo Perrone
- b Division of Oncology , I.R.C.C.S. Istituti Clinici Scientifici Maugeri , Pavia , Italy
| | - Eugenia Bosco
- a Division of Translational Oncology , I.R.C.C.S. Istituti Clinici Scientifici Maugeri , Pavia , Italy
| | - Giuseppe Saltalamacchia
- a Division of Translational Oncology , I.R.C.C.S. Istituti Clinici Scientifici Maugeri , Pavia , Italy
| | - Alessandra Mosca
- c Medical Oncology , Ospedale Maggiore della Carità , Novara , Italy
| | - Mimma Rizzo
- a Division of Translational Oncology , I.R.C.C.S. Istituti Clinici Scientifici Maugeri , Pavia , Italy
| | - Camillo Porta
- a Division of Translational Oncology , I.R.C.C.S. Istituti Clinici Scientifici Maugeri , Pavia , Italy.,d Department of Internal Medicine , University of Pavia , Pavia , Italy
| |
Collapse
|
3157
|
Hahn AW, Nussenzveig RH, Maughan BL, Agarwal N. Cell-free Circulating Tumor DNA (ctDNA) in Metastatic Renal Cell Carcinoma (mRCC): Current Knowledge and Potential Uses. KIDNEY CANCER 2019. [DOI: 10.3233/kca-180048] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Andrew W. Hahn
- Department of Internal Medicine, Division of Oncology, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Roberto H. Nussenzveig
- Department of Internal Medicine, Division of Oncology, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Benjamin L. Maughan
- Department of Internal Medicine, Division of Oncology, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Neeraj Agarwal
- Department of Internal Medicine, Division of Oncology, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
3158
|
Bergerot PG, Bergerot CD, Philip EJ, Meza L, Dizman N, Hsu J, Pal SK. Targeted Therapy and Immunotherapy: Effect of Body Mass Index on Clinical Outcomes in Patients Diagnosed with Metastatic Renal Cell Carcinoma. KIDNEY CANCER 2019. [DOI: 10.3233/kca-180047] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Affiliation(s)
- Paulo Gustavo Bergerot
- Department of Medical Oncology & Experimental Therapeutics, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Cristiane Decat Bergerot
- Department of Medical Oncology & Experimental Therapeutics, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | | | - Luis Meza
- Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Nazli Dizman
- Department of Medical Oncology & Experimental Therapeutics, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - JoAnn Hsu
- Department of Medical Oncology & Experimental Therapeutics, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Sumanta Kumar Pal
- Department of Medical Oncology & Experimental Therapeutics, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| |
Collapse
|
3159
|
Reed JP, Posadas EM, Figlin RA. Developments in the use of tyrosine kinase inhibitors in the treatment of renal cell carcinoma. Expert Rev Anticancer Ther 2019; 19:259-271. [PMID: 30669895 DOI: 10.1080/14737140.2019.1573678] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
INTRODUCTION Renal cell carcinoma (RCC) is among the most commonly diagnosed solid malignancies, but until recently there were few systemic treatment options for advanced disease. Since 2005, the treatment landscape has been transformed by the development of several novel systemic therapies. In particular, tyrosine kinase inhibitors (TKIs) targeting the vascular endothelial growth factor (VEGF) pathway have been instrumental in improving outcomes in patients with metastatic disease. Areas covered: The armamentarium of TKIs available for the treatment of RCC has expanded in recent years. The most active area of research at this time is the development of treatment regimens combining newer-generation TKIs and immune checkpoint inhibitors. Emerging data point to a role for combination therapy in the frontline management of advanced RCC. Other ongoing areas of research include the use of TKIs in the adjuvant setting and the role of cytoreductive nephrectomy within a changing treatment landscape. Expert opinion: Although TKIs and immune checkpoint inhibitors have incrementally improved outcomes for patients with advanced RCC, long-term survival remains poor. The development of regimens combining these agents represents the next step in the evolution of the field. For the clinician, this will offer exciting possibilities and novel challenges.
Collapse
Affiliation(s)
- Jarred P Reed
- a Department of Medicine, Division of Hematology and Oncology , Cedars-Sinai Medical Center , Los Angeles , CA , USA
| | - Edwin M Posadas
- a Department of Medicine, Division of Hematology and Oncology , Cedars-Sinai Medical Center , Los Angeles , CA , USA
| | - Robert A Figlin
- a Department of Medicine, Division of Hematology and Oncology , Cedars-Sinai Medical Center , Los Angeles , CA , USA
| |
Collapse
|
3160
|
Mouillet G, Fritzsch J, Paget-Bailly S, Pozet A, Es-Saad I, Meurisse A, Vernerey D, Mouyabi K, Berthod D, Bonnetain F, Anota A, Thiery-Vuillemin A. Health-related quality of life assessment for patients with advanced or metastatic renal cell carcinoma treated with a tyrosine kinase inhibitor using electronic patient-reported outcomes in daily clinical practice (QUANARIE trial): study protocol. Health Qual Life Outcomes 2019; 17:25. [PMID: 30717745 PMCID: PMC6360763 DOI: 10.1186/s12955-019-1085-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 01/07/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Two main therapies, pazopanib and sunitinib, are used in the first-line setting for metastatic renal cell carcinoma (mRCC). These two tyrosine kinase inhibitors (TKI) are equally effective in terms of survival; however, they frequently induce adverse events. In this setting, Health-Related Quality of life (HRQoL) is a key element in the choice between these two treatments and the evaluation of treatment effectiveness. It could be of interest to evaluate HRQoL in daily clinical practice to aid adequate therapy choice and management. Currently, the development of information and communication technology may allow HRQoL monitoring in routine practice. The objective of the QUANARIE study is to evaluate the use of HRQoL assessment in daily clinical practice for patients with mRCC treated with TKI using electronic patient-reported outcomes (e-PRO). The present article describes the key elements of the study protocol. METHODS The QUANARIE study is an interventional, prospective, multicentre trial. Patients diagnosed with mRCC initiating sunitinib or pazopanib treatment will be invited to complete the EORTC QLQ-C30 questionnaire, nine additional questions from the EORTC items library, and the EuroQoL EQ-5D, prior to each visit with the physician. Questionnaires will be completed by patients using tablets and/or computer terminals via the e-PRO software. The physician will have real-time access to a visual summary of the HRQoL evaluation. The primary objective is to assess the proportion of patients having good compliance with Routine Electronic Monitoring of HRQoL (REMOQOL) during the first 12 months. Physicians' satisfaction with REMOQOL will be assessed as a secondary objective. We hypothesise that 80% of patients having good compliance with REMOQOL would be meaningful. A sample size of 56 patients would be needed. DISCUSSION The results of this study will show whether REMOQOL is feasible on a large scale and whether patients are receptive to this new practice. This study will also determine how real-time multidimensional evaluation of patient perception can help physicians in their daily practice and how they used it in conjunction with other clinical information to manage patient care. TRIAL REGISTRATION ClinicalTrials.gov; Identifier: NCT03062410 ; First Posted: February 23, 2017; Last Update Posted: August 9, 2017.
Collapse
Affiliation(s)
- Guillaume Mouillet
- Department of Medical Oncology, University Hospital of Besançon, Boulevard Fleming, F-25000 Besançon, France
- Methodological and Quality of Life Unit, University Hospital of Besançon, F-25000 Besançon, France
- INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, University Bourgogne Franche-Comté, F-25000 Besançon, France
| | - Joëlle Fritzsch
- Methodological and Quality of Life Unit, University Hospital of Besançon, F-25000 Besançon, France
- INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, University Bourgogne Franche-Comté, F-25000 Besançon, France
| | - Sophie Paget-Bailly
- Methodological and Quality of Life Unit, University Hospital of Besançon, F-25000 Besançon, France
- INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, University Bourgogne Franche-Comté, F-25000 Besançon, France
| | - Astrid Pozet
- Methodological and Quality of Life Unit, University Hospital of Besançon, F-25000 Besançon, France
- INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, University Bourgogne Franche-Comté, F-25000 Besançon, France
| | - Ikram Es-Saad
- Methodological and Quality of Life Unit, University Hospital of Besançon, F-25000 Besançon, France
- INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, University Bourgogne Franche-Comté, F-25000 Besançon, France
| | - Aurelia Meurisse
- Methodological and Quality of Life Unit, University Hospital of Besançon, F-25000 Besançon, France
- INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, University Bourgogne Franche-Comté, F-25000 Besançon, France
| | - Dewi Vernerey
- Methodological and Quality of Life Unit, University Hospital of Besançon, F-25000 Besançon, France
- INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, University Bourgogne Franche-Comté, F-25000 Besançon, France
| | - Kristina Mouyabi
- Clinical Research and Innovation Office, University Hospital of Besançon, F-25000 Besançon, France
| | - Diane Berthod
- Department of Medical Oncology, University Hospital of Besançon, Boulevard Fleming, F-25000 Besançon, France
| | - Franck Bonnetain
- Methodological and Quality of Life Unit, University Hospital of Besançon, F-25000 Besançon, France
- INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, University Bourgogne Franche-Comté, F-25000 Besançon, France
| | - Amélie Anota
- Methodological and Quality of Life Unit, University Hospital of Besançon, F-25000 Besançon, France
- INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, University Bourgogne Franche-Comté, F-25000 Besançon, France
- French National Platform Quality of Life and Cancer, Besançon, France
| | - Antoine Thiery-Vuillemin
- Department of Medical Oncology, University Hospital of Besançon, Boulevard Fleming, F-25000 Besançon, France
- INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, University Bourgogne Franche-Comté, F-25000 Besançon, France
| |
Collapse
|
3161
|
The efficacy and safety of sunitinib given on an individualised schedule as first-line therapy for metastatic renal cell carcinoma: A phase 2 clinical trial. Eur J Cancer 2019; 108:69-77. [DOI: 10.1016/j.ejca.2018.12.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 11/24/2018] [Accepted: 12/09/2018] [Indexed: 11/23/2022]
|
3162
|
CheckMate 214 patient-reported outcomes: listening to our patients. Lancet Oncol 2019; 20:179-180. [DOI: 10.1016/s1470-2045(18)30790-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Accepted: 10/17/2018] [Indexed: 12/21/2022]
|
3163
|
Reinhorn D, Sarfaty M, Leshno M, Moore A, Neiman V, Rosenbaum E, Goldstein DA. A Cost-Effectiveness Analysis of Nivolumab and Ipilimumab Versus Sunitinib in First-Line Intermediate- to Poor-Risk Advanced Renal Cell Carcinoma. Oncologist 2019; 24:366-371. [PMID: 30710066 DOI: 10.1634/theoncologist.2018-0656] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 12/28/2018] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND The treatment paradigm of advanced renal cell carcinoma (RCC) has changed rapidly in recent years. In first-line treatment of intermediate- to poor-risk patients, the CheckMate 214 study demonstrated a significant survival advantage for nivolumab and ipilimumab versus sunitinib. The high cost of combined immune-modulating agents warrants an understanding of the combination's value by considering both efficacy and cost. The objective of this study was to estimate the cost-effectiveness of nivolumab and ipilimumab compared with sunitinib for first-line treatment of intermediate- to poor-risk advanced RCC from the U.S. payer perspective. MATERIALS AND METHODS A Markov model was developed to compare the costs and effectiveness of nivolumab and ipilimumab with those of sunitinib in the first-line treatment of intermediate- to poor-risk advanced RCC. Health outcomes were measured in life-years and quality-adjusted life-years (QALYs). Drug costs were based on Medicare reimbursement rates in 2017. We extrapolated survival beyond the trial closure using Weibull distribution. Model robustness was addressed in univariable and probabilistic sensitivity analyses. RESULTS The total mean cost per-patient of nivolumab and ipilimumab versus sunitinib was $292,308 and $169,287, respectfully. Nivolumab and ipilimumab generated a gain of 0.978 QALYs over sunitinib. The incremental cost-effectiveness ratio (ICER) for nivolumab and ipilimumab was $125,739/QALY versus sunitinib. CONCLUSION Our analysis established that the base case ICER in the model for nivolumab and ipilimumab versus sunitinib is below what some would consider the upper limit of the theoretical willingness-to-pay threshold in the U.S. ($150,000/QALY) and is thus estimated to be cost-effective. IMPLICATIONS FOR PRACTICE This article assessed the cost-effectiveness of nivolumab and ipilimumab versus sunitinib for treatment of patients with intermediate- to poor-risk metastatic kidney cancer, from the U.S. payer perspective. It would cost $125,739 to gain 1 quality-adjusted life-year with nivolumab and ipilimumab versus sunitinib in these patients.
Collapse
Affiliation(s)
- Daniel Reinhorn
- Institute of Oncology, Davidoff Cancer Center, Rabin Medical Center, Petah Tikva, Israel
| | - Michal Sarfaty
- Institute of Oncology, Davidoff Cancer Center, Rabin Medical Center, Petah Tikva, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Moshe Leshno
- Coller School of Management, Tel Aviv University, Tel Aviv, Israel
| | - Assaf Moore
- Institute of Oncology, Davidoff Cancer Center, Rabin Medical Center, Petah Tikva, Israel
| | - Victoria Neiman
- Institute of Oncology, Davidoff Cancer Center, Rabin Medical Center, Petah Tikva, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Eli Rosenbaum
- Institute of Oncology, Davidoff Cancer Center, Rabin Medical Center, Petah Tikva, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Daniel A Goldstein
- Institute of Oncology, Davidoff Cancer Center, Rabin Medical Center, Petah Tikva, Israel
- Department of Health Policy and Management, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, North Carolina, USA
| |
Collapse
|
3164
|
Lemiale V, Meert AP, Vincent F, Darmon M, Bauer PR, Van de Louw A, Azoulay E. Severe toxicity from checkpoint protein inhibitors: What intensive care physicians need to know? Ann Intensive Care 2019; 9:25. [PMID: 30707321 PMCID: PMC6358632 DOI: 10.1186/s13613-019-0487-x] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Accepted: 01/12/2019] [Indexed: 12/13/2022] Open
Abstract
Checkpoint protein inhibitor antibodies (CPI), including cytotoxic T-lymphocyte-associated antigen 4 inhibitors (ipilimumab, tremelimumab) and the programmed cell death protein 1 pathway/programmed cell death protein 1 ligand inhibitors (pembrolizumab, nivolumab, durvalumab, atezolizumab), have entered routine practice for the treatment of many cancers. They improve the outcome for many cancers, and more patients will be treated with CPI in the future. Although CPI can lead to adverse events (AE) less frequently than for chemotherapy, their use can require intensive care unit admission in case of severe immune-related adverse events (IrAE). Moreover, some of these events, particularly late events, are poorly documented, so a high level of suspicion should be maintained for patients receiving CPI. Intensivists should be aware in general of the known complications and appropriate management of these AE. Nevertheless, a multidisciplinary collaboration remains essential for their diagnosis and management. This review described the most severe complications related to CPI.
Collapse
Affiliation(s)
- Virginie Lemiale
- Medical Intensive Care Unit, APHP, Hôpital Saint-Louis, Paris, France.
| | - Anne-Pascale Meert
- Soins Intensifs et urgences oncologiques, Institut Jules Bordet (ULB), Brussels, Belgium
| | - François Vincent
- Medical Surgical ICU, GHIC Le Raincy-Montfermeil, 93370, Montfermeil, France
| | - Michael Darmon
- Medical Intensive Care Unit, APHP, Hôpital Saint-Louis, Paris, France.,ECSTRA, Saint Louis SBIM, APHP, Paris, France
| | | | - Andry Van de Louw
- Division of Pulmonary and Critical Care Medicine, Penn State University College of Medicine, Hershey, PA, USA
| | - Elie Azoulay
- Medical Intensive Care Unit, APHP, Hôpital Saint-Louis, Paris, France.,ECSTRA, Saint Louis SBIM, APHP, Paris, France
| | | |
Collapse
|
3165
|
Auvray M, Auclin E, Barthelemy P, Bono P, Kellokumpu-Lehtinen P, Gross-Goupil M, De Velasco G, Powles T, Mouillet G, Vano YA, Gravis G, Mourey L, Priou F, Rolland F, Escudier B, Albiges L. Second-line targeted therapies after nivolumab-ipilimumab failure in metastatic renal cell carcinoma. Eur J Cancer 2019; 108:33-40. [DOI: 10.1016/j.ejca.2018.11.031] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 11/07/2018] [Accepted: 11/19/2018] [Indexed: 10/27/2022]
|
3166
|
Isono T, Chano T, Yoshida T, Makino A, Ishida S, Suzaki M, Kageyama S, Kawauchi A, Yonese J, Yuasa T. ADP-ribosylation factor-like 4C is a predictive biomarker of poor prognosis in patients with renal cell carcinoma. Am J Cancer Res 2019; 9:415-423. [PMID: 30906638 PMCID: PMC6405968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Accepted: 01/23/2019] [Indexed: 06/09/2023] Open
Abstract
Renal cell carcinoma (RCC) has the high mortality rate among urological malignancies. The development of RCC cannot be effectively reduced by molecular targeted therapies based on nutrient deprivation, such as inhibition of tumor angiogenesis. The objective of this study was to identify predictive biomarkers of poor prognosis and therapeutic molecular targets in patients with RCC. Two independent cohorts were analyzed in the present study. Global transcriptomics were used in the first cohort (43 patients with RCC) to identify biomarker genes. Each identified biomarker was subsequently analyzed using immunohistochemistry in the second cohort (97 patients with RCC). Following transcriptomics, biomarkers were evaluated using receiver operating characteristic curve analysis. Predictive accuracy for poor survivals was assessed using the log-rank test and Cox multivariate analysis. Global transcriptomic analysis in the first cohort focusing on cases with survival periods <2 years after initial diagnosis of metastasis detected seven overexpressed genes, which correlated with poor prognosis. The ADP-ribosylation factor-like 4C (ARL4C) exhibited the best accuracy in the receiver operating characteristic curve analysis and predicted poor survival in the first cohort (log-rank test, P<0.001; Cox multivariate analysis, hazard ratio =167, P=0.005). In the second cohort, the expression of ARL4C was semi-quantitatively evaluated through immunohistochemistry. Twenty-seven cases showed high levels of ARL4C, confirming a significant association with shorter survivals (log-rank test, P<0.001; Cox multivariate analysis, hazard ratio =9.41, P=0.004). ARL4C was shown to be a predictive biomarker for poor prognosis in patients with RCC and may be a novel target in the treatment of RCC.
Collapse
Affiliation(s)
- Takahiro Isono
- Central Research Laboratory, Shiga University of Medical ScienceOtsu, Shiga 520-2192, Japan
| | - Tokuhiro Chano
- Department of Clinical Laboratory Medicine, Shiga University of Medical ScienceOtsu, Shiga 520-2192, Japan
| | - Tetsuya Yoshida
- Department of Urology, Shiga University of Medical ScienceOtsu, Shiga 520-2192, Japan
| | - Ai Makino
- Central Research Laboratory, Shiga University of Medical ScienceOtsu, Shiga 520-2192, Japan
| | - Shohei Ishida
- Central Research Laboratory, Shiga University of Medical ScienceOtsu, Shiga 520-2192, Japan
| | - Masafumi Suzaki
- Central Research Laboratory, Shiga University of Medical ScienceOtsu, Shiga 520-2192, Japan
| | - Susumu Kageyama
- Department of Urology, Shiga University of Medical ScienceOtsu, Shiga 520-2192, Japan
| | - Akihiro Kawauchi
- Department of Urology, Shiga University of Medical ScienceOtsu, Shiga 520-2192, Japan
| | - Junji Yonese
- Department of Urology, Cancer Institute Hospital, Japanese Foundation for Cancer ResearchTokyo 135-8550, Japan
| | - Takeshi Yuasa
- Department of Urology, Cancer Institute Hospital, Japanese Foundation for Cancer ResearchTokyo 135-8550, Japan
| |
Collapse
|
3167
|
Nakayama T, Kitano S. Immunotherapy for genitourinary tumors. Int J Urol 2019; 26:326-333. [PMID: 30710374 DOI: 10.1111/iju.13902] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 12/04/2018] [Indexed: 12/14/2022]
Abstract
The present review provides an update about the major achievements and recent advances of immunotherapy in renal cell carcinoma, urothelial carcinoma, and prostate cancer. Although the treatment strategy for renal cell carcinoma and urothelial carcinoma includes traditional cancer immunotherapies, such as interleukin-2 and interferon-alfa, the clinical outcomes of these therapies are unsatisfactory. In recent years, the development of immune checkpoint inhibitors has drastically changed the treatment strategy for various cancers, including genitourinary cancer. The present review summarizes the approved cancer immunotherapies for renal cell carcinoma, urothelial carcinoma and prostate cancer. Furthermore, we review the response evaluation and biomarkers for immune checkpoint inhibitors with a distinctive mode of action that is different from cytotoxic agents. Finally, future perspectives for cancer immunotherapy are discussed.
Collapse
Affiliation(s)
- Takayuki Nakayama
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo, Japan
| | - Shigehisa Kitano
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo, Japan
| |
Collapse
|
3168
|
Solanki AA, Bossi A, Efstathiou JA, Lock D, Mondini M, Ramapriyan R, Welsh J, Kang J. Combining Immunotherapy with Radiotherapy for the Treatment of Genitourinary Malignancies. Eur Urol Oncol 2019; 2:79-87. [PMID: 30929848 DOI: 10.1016/j.euo.2018.09.013] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 09/17/2018] [Accepted: 09/26/2018] [Indexed: 11/15/2022]
Abstract
CONTEXT Immunotherapy drugs, particularly checkpoint inhibitors, have recently been approved by the Food and Drug Administration for various malignancies. Preclinical and early clinical data show that combining these agents with radiotherapy may produce an even more potent antitumor effect in the treatment of cancer. OBJECTIVE To describe the rationale, available data, and emerging data on the use of combined immunotherapy and radiation therapy in the setting of genitourinary (GU) malignancies. EVIDENCE ACQUISITION We performed a search of primary studies from PubMed/Medline that included combinations of the search terms "radiation therapy," "radiotherapy," "abscopal effect," "immunotherapy," "combined," and "combination." EVIDENCE SYNTHESIS Preclinical and clinical data support both immune-stimulating and immune-suppressing effects of radiotherapy. Preclinical and clinical studies investigating the combination of radiotherapy with immunotherapy, primarily in the setting of non-GU malignancies, have suggested efficacy and tolerability. Early randomized trials combining radiotherapy and immunotherapy have demonstrated success in lung cancer. Although a trial investigating combined immunotherapy and radiotherapy use for prostate cancer did not clearly improve survival, trials are ongoing in multiple GU malignancies to identify synergy between immunotherapy and radiotherapy. Several practical and technical questions remain about the optimal combination of radiotherapy and immunotherapy. CONCLUSIONS Preclinical and clinical trials show that the combination of the immunotherapy and radiation therapy has the potential to provide a synergistic effect in treating cancer, including GU malignancies, although more work is needed to uncover the mechanism and determine the optimal delivery of this treatment. PATIENT SUMMARY This paper reviews evidence that immunotherapy drugs can be given together with radiation therapy to improve outcomes in cancers of the genitourinary tract. We find promising initial results and raise important questions that need to be answered before this type of treatment can be utilized successfully.
Collapse
Affiliation(s)
- Abhishek A Solanki
- Department of Radiation Oncology, Loyola University Chicago Stritch School of Medicine, Maywood, IL, USA.
| | - Alberto Bossi
- Department of Radiation Oncology, Gustave Roussy Villejuif, France
| | - Jason A Efstathiou
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Derrick Lock
- Department of Radiation Oncology, Loyola University Chicago Stritch School of Medicine, Maywood, IL, USA
| | - Michele Mondini
- Gustave Roussy, Université Paris-Saclay, Villejuif, France; Labex LERMIT, SIRIC SOCRATE, Villejuif, France
| | - Rishab Ramapriyan
- Department of Radiation Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - James Welsh
- Department of Radiation Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - Josephine Kang
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
3169
|
Pignot G, Loriot Y, Kamat AM, Shariat SF, Plimack ER. Effect of Immunotherapy on Local Treatment of Genitourinary Malignancies. Eur Urol Oncol 2019; 2:355-364. [PMID: 31277773 DOI: 10.1016/j.euo.2019.01.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 12/09/2018] [Accepted: 01/07/2019] [Indexed: 12/28/2022]
Abstract
CONTEXT Management of metastatic genitourinary malignancies has recently been transformed through the use of immune checkpoint inhibitors. The best way to integrate them into local treatment paradigms is still under investigation. OBJECTIVE To systematically evaluate evidence regarding the use of immunotherapy in the treatment of local disease, in both the perioperative and the metastatic setting. EVIDENCE ACQUISITION We performed a critical review of PubMed and ClinicalTrials.gov according to the Preferred Reporting Items for Systematic Review and Meta-analyses (PRISMA) statement. Prospective and retrospective studies between 2011 and 2018 were included. Twenty-four publications were selected for inclusion, including 10 on urothelial carcinoma, seven on renal cell carcinoma, six on prostate cancer, and one on germ-cell cancer. EVIDENCE SYNTHESIS Prospective early-phase trials investigating neoadjuvant immunotherapy prior to cystectomy in urothelial carcinoma suggest a high rate of pathological complete response, from 29% with atezolizumab to 39.5% with pembrolizumab. Several neoadjuvant and adjuvant trials are still ongoing in bladder, renal, and prostate cancers, before or after surgery. The combination of immunotherapy and radiotherapy is being explored and could offer an interesting strategy for definitive treatment modality with curative intent. Finally, in metastatic disease, delayed local treatment could be discussed after immunotherapy in selected patients with an excellent radiographic response. CONCLUSIONS Little evidence exists on the oncological impact of immunotherapy on the local treatment of genitourinary malignancies, but preliminary results are encouraging and many prospective trials are ongoing. PATIENT SUMMARY In this study, we review recent advances in immunotherapy and its role in local treatment. Immunotherapy is evaluated before or after surgery, or in combination with radiotherapy for localized disease. Ongoing trials will bring clarity on the local downstaging effect of immunotherapy and its association with oncological and functional outcomes.
Collapse
Affiliation(s)
- Géraldine Pignot
- Institut Paoli-Calmettes, Chirurgie Oncologique 2, Marseille, France.
| | - Yohann Loriot
- Institut Gustave Roussy, Institut National de la Santé et de la Recherche Médicale U981, University of Paris Saclay, Villejuif, France
| | - Ashish M Kamat
- Department of Urology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Elizabeth R Plimack
- Department of Hematology/Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA
| |
Collapse
|
3170
|
Green YS, Sargis T, Reichert EC, Rudasi E, Fuja D, Jonasch E, Koh MY. Hypoxia-Associated Factor (HAF) Mediates Neurofibromin Ubiquitination and Degradation Leading to Ras-ERK Pathway Activation in Hypoxia. Mol Cancer Res 2019; 17:1220-1232. [PMID: 30705246 DOI: 10.1158/1541-7786.mcr-18-1080] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 01/07/2019] [Accepted: 01/24/2019] [Indexed: 01/05/2023]
Abstract
Low oxygen or hypoxia is a feature of all solid tumors and has been associated with aggressive disease. Here, we describe a novel mechanism for the hypoxia-dependent degradation of the Ras-GTPase-activating protein neurofibromin, by hypoxia-associated factor (HAF). We have previously characterized HAF as an oxygen-independent ubiquitin ligase for HIF-1α. Here, we show that HAF promotes neurofibromin ubiquitination and degradation independently of oxygen and pVHL, resulting in Ras-ERK pathway activation. Hypoxia enhanced HAF:neurofibromin binding independently of HAF-SUMOylation, whereas HAF knockdown increased neurofibromin levels primarily in hypoxia, supporting the role of HAF as a hypoxia-specific neurofibromin regulator. HAF overexpression increased p-ERK levels and promoted resistance of clear cell kidney cancer (ccRCC) cells to sorafenib and sunitinib in both normoxia and hypoxia. However, a greater-fold increase in sorafenib/sunitinib resistance was observed during hypoxia, particularly in pVHL-deficient cells. Intriguingly, HAF-mediated resistance was HIF-2α-dependent in normoxia, but HIF-2α-independent in hypoxia indicating two potential mechanisms of HAF-mediated resistance: a HIF-2α-dependent pathway dominant in normoxia, and the direct activation of the Ras-ERK pathway through neurofibromin degradation dominant in hypoxia. Patients with ccRCC with high HAF transcript or protein levels showed significantly decreased overall survival compared with those with low HAF. Thus, we establish a novel, nonmutational pathway of neurofibromin inactivation through hypoxia-induced HAF-mediated degradation, leading to Ras-ERK activation and poor prognosis in ccRCC. IMPLICATIONS: We describe a novel mechanism of neurofibromin degradation induced by hypoxia that leads to activation of the prooncogenic Ras-ERK pathway and resistance to therapy.
Collapse
Affiliation(s)
- Yangsook Song Green
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, Utah
| | - Timothy Sargis
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | | | - Eleanor Rudasi
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, Utah
| | - Daniel Fuja
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, Utah
| | - Eric Jonasch
- Department of Genitourinary Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mei Yee Koh
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, Utah.
| |
Collapse
|
3171
|
Ikarashi D, Nakamura Y, Shimodate H, Usui Y, Ujiie T, Obara W. Complete response to perioperative treatment using nivolumab for metastatic renal cell carcinoma: A case report. Urol Case Rep 2019; 24:100839. [PMID: 31211050 PMCID: PMC6562266 DOI: 10.1016/j.eucr.2019.100839] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Revised: 01/25/2019] [Accepted: 01/29/2019] [Indexed: 12/23/2022] Open
Abstract
We report a case of 59-year-old woman who has multiple lung metastases with renal cell carcinoma (RCC). She received neoadjuvant therapy using nivolumab following sunitinib. Thereafter, we performed cytoreductive nephrectomy and subsequently administered nivolumab. We also found a high expression of PD-L1 in tumor cell and infiltration of lymphocytes with CD8 expression by immunohistochemistry. A complete response was achieved 4 months after surgery. A perioperative treatment using nivolumab might be useful treatment for metastatic RCC.
Collapse
Affiliation(s)
- Daiki Ikarashi
- Department of Urology, Iwate Medical University School of Medicine, 19-1, Uchimaru, Morioka-shi, Iwate, 020-8505, Japan.,Department of Urology, Iwate Prefectural Ofunato Hospital, 10-1, Yamaumagoe, Ofunato-shi, Iwate, 022-8512, Japan
| | - Yasuyuki Nakamura
- Department of Pathology, Iwate Prefectural Ofunato Hospital, 10-1, Yamaumagoe, Ofunato-shi, Iwate, 022-8512, Japan
| | - Hitoshi Shimodate
- Department of Urology, Iwate Medical University School of Medicine, 19-1, Uchimaru, Morioka-shi, Iwate, 020-8505, Japan
| | - Yoshitaka Usui
- Department of Urology, Iwate Prefectural Ofunato Hospital, 10-1, Yamaumagoe, Ofunato-shi, Iwate, 022-8512, Japan
| | - Takashi Ujiie
- Department of Urology, Iwate Prefectural Ofunato Hospital, 10-1, Yamaumagoe, Ofunato-shi, Iwate, 022-8512, Japan
| | - Wataru Obara
- Department of Urology, Iwate Medical University School of Medicine, 19-1, Uchimaru, Morioka-shi, Iwate, 020-8505, Japan
| |
Collapse
|
3172
|
Thanindratarn P, Dean DC, Nelson SD, Hornicek FJ, Duan Z. Advances in immune checkpoint inhibitors for bone sarcoma therapy. J Bone Oncol 2019; 15:100221. [PMID: 30775238 PMCID: PMC6365405 DOI: 10.1016/j.jbo.2019.100221] [Citation(s) in RCA: 103] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 01/24/2019] [Accepted: 01/24/2019] [Indexed: 12/22/2022] Open
Abstract
Bone sarcomas are a collection of sporadic malignancies of mesenchymal origin. The most common subtypes include osteosarcoma, Ewing sarcoma, chondrosarcoma, and chordoma. Despite the use of aggressive treatment protocols consisting of extensive surgical resection, chemotherapy, and radiotherapy, outcomes have not significantly improved over the past few decades for osteosarcoma or Ewing sarcoma patients. In addition, chondrosarcoma and chordoma are resistant to both chemotherapy and radiation therapy. There is, therefore, an urgent need to elucidate which novel new therapies may affect bone sarcomas. Emerging checkpoint inhibitors have generated considerable attention for their clinical success in a variety of human cancers, which has led to works assessing their potential in bone sarcoma management. Here, we review the recent advances of anti-PD-1/PD-L1 and anti-CTLA-4 blockade as well as other promising new immune checkpoint targets for their use in bone sarcoma therapy.
Collapse
Affiliation(s)
- Pichaya Thanindratarn
- Department of Orthopedic Surgery, Sarcoma Biology Laboratory, David Geffen School of Medicine, University of California, 615 Charles E. Young. Dr. South, Los Angeles, CA 90095, USA
- Department of Orthopedic Surgery, Chulabhorn hospital, HRH Princess Chulabhorn College of Medical Science, Bangkok, Thailand
| | - Dylan C. Dean
- Department of Orthopedic Surgery, Sarcoma Biology Laboratory, David Geffen School of Medicine, University of California, 615 Charles E. Young. Dr. South, Los Angeles, CA 90095, USA
| | - Scott D. Nelson
- Department of Pathology, University of California, Los Angeles, CA, USA
| | - Francis J. Hornicek
- Department of Orthopedic Surgery, Sarcoma Biology Laboratory, David Geffen School of Medicine, University of California, 615 Charles E. Young. Dr. South, Los Angeles, CA 90095, USA
| | - Zhenfeng Duan
- Department of Orthopedic Surgery, Sarcoma Biology Laboratory, David Geffen School of Medicine, University of California, 615 Charles E. Young. Dr. South, Los Angeles, CA 90095, USA
- Corresponding author.
| |
Collapse
|
3173
|
Grünwald V, Powles T, Choueiri TK, Hutson TE, Porta C, Eto M, Sternberg CN, Rha SY, He CS, Dutcus CE, Smith A, Dutta L, Mody K, Motzer RJ. Lenvatinib plus everolimus or pembrolizumab versus sunitinib in advanced renal cell carcinoma: study design and rationale. Future Oncol 2019; 15:929-941. [PMID: 30689402 DOI: 10.2217/fon-2018-0745] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
AIM Lenvatinib plus everolimus is approved for the treatment of advanced renal cell carcinoma (RCC) after one prior vascular endothelial growth factor-targeted therapy. Lenvatinib plus pembrolizumab demonstrated promising antitumor activity in a Phase I/II trial of RCC. METHODS We describe the rationale and design of the CLEAR study, a three-arm Phase III trial comparing lenvatinib plus everolimus and lenvatinib plus pembrolizumab versus sunitinib monotherapy for first-line treatment of RCC. Eligible patients must have advanced clear cell RCC and must not have received any prior systemic anticancer therapy. The primary end point is progression-free survival; secondary end points include objective response rate, overall survival, safety, health-related quality of life and pharmacokinetics. Biomarker evaluations are included as exploratory end points.
Collapse
Affiliation(s)
- Viktor Grünwald
- Clinic for Hematology, Hemostasis, Oncology and Stem Cell Transplantation, University Hospital Essen, Essen, Germany
| | - Thomas Powles
- Experimental Cancer Medicine, Barts Cancer Institute, London, UK
| | - Toni K Choueiri
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Thomas E Hutson
- Urologic Oncology Program, Baylor University Medical Center, Dallas, TX, USA
| | - Camillo Porta
- University of Pavia & Division of Translational Oncology, IRCCS Istituti Clinici Scientifici Maugeri, Pavia, Italy
| | - Masatoshi Eto
- Department of Urology, Kyushu University, Fukuoka, Japan
| | - Cora N Sternberg
- Weill Cornell Medicine, New York-Presbyterian, New York, NY, USA
| | - Sun Young Rha
- Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, South Korea
| | | | | | | | | | | | - Robert J Motzer
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
3174
|
Zhang M, Yang J, Hua W, Li Z, Xu Z, Qian Q. Monitoring checkpoint inhibitors: predictive biomarkers in immunotherapy. Front Med 2019; 13:32-44. [PMID: 30680606 DOI: 10.1007/s11684-018-0678-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 11/16/2018] [Indexed: 12/19/2022]
Abstract
Immunotherapy has become the fourth cancer therapy after surgery, chemotherapy, and radiotherapy. In particular, immune checkpoint inhibitors are proved to be unprecedentedly in increasing the overall survival rates of patients with refractory cancers, such as advanced melanoma, non-small cell lung cancer, and renal cell carcinoma. However, inhibitor therapies are only effective in a small proportion of patients with problems, such as side effects and high costs. Therefore, doctors urgently need reliable predictive biomarkers for checkpoint inhibitor therapies to choose the optimal therapies. Here, we review the biomarkers that can serve as potential predictors of the outcomes of immune checkpoint inhibitor treatment, including tumor-specific profiles and tumor microenvironment evaluation and other factors.
Collapse
Affiliation(s)
- Min Zhang
- ShangHai Cell Therapy Group Co., Ltd., Shanghai, 201805, China
| | - Jingwen Yang
- ShangHai Cell Therapy Group Co., Ltd., Shanghai, 201805, China
| | - Wenjing Hua
- ShangHai Cell Therapy Group Co., Ltd., Shanghai, 201805, China
| | - Zhong Li
- ShangHai Cell Therapy Group Co., Ltd., Shanghai, 201805, China
| | - Zenghui Xu
- ShangHai Cell Therapy Group Co., Ltd., Shanghai, 201805, China.
| | - Qijun Qian
- ShangHai Cell Therapy Group Co., Ltd., Shanghai, 201805, China.
| |
Collapse
|
3175
|
Seto T, Sam D, Pan M. Mechanisms of Primary and Secondary Resistance to Immune Checkpoint Inhibitors in Cancer. Med Sci (Basel) 2019; 7:E14. [PMID: 30678257 PMCID: PMC6410194 DOI: 10.3390/medsci7020014] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 01/10/2019] [Accepted: 01/15/2019] [Indexed: 12/14/2022] Open
Abstract
Immune checkpoint inhibitors (ICPis) have revolutionized cancer therapy with broad activities against a wide range of malignancies. However, in many malignancies their efficacy remains limited due to the primary resistance. Furthermore, a high percentage of patients develop progression due to the secondary resistance even after obtaining a response or achieving a stable disease. In this review, we will discuss the mechanisms that underlie the primary and secondary resistance to ICPis in cancer immunotherapy and provide an overview to impart a broad understanding of the critical issues that are encountered in clinical oncology practice.
Collapse
Affiliation(s)
| | - Danny Sam
- Internal Medicine Residency Program.
| | - Minggui Pan
- Internal Medicine Residency Program.
- Department of Oncology and Hematology, Kaiser Permanente, Santa Clara, CA 95051, USA; and Kaiser Permanente Division of Research, Oakland, CA 94612, USA.
| |
Collapse
|
3176
|
Kucharz J, Dumnicka P, Kusnierz-Cabala B, Demkow T, Wiechno P. The correlation between the incidence of adverse events and progression-free survival in patients treated with cabozantinib for metastatic renal cell carcinoma (mRCC). Med Oncol 2019; 36:19. [DOI: 10.1007/s12032-018-1239-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 12/17/2018] [Indexed: 12/30/2022]
|
3177
|
Ndiaye PD, Dufies M, Giuliano S, Douguet L, Grépin R, Durivault J, Lenormand P, Glisse N, Mintcheva J, Vouret-Craviari V, Mograbi B, Wurmser M, Ambrosetti D, Rioux-Leclercq N, Maire P, Pagès G. VEGFC acts as a double-edged sword in renal cell carcinoma aggressiveness. Am J Cancer Res 2019; 9:661-675. [PMID: 30809300 PMCID: PMC6376471 DOI: 10.7150/thno.27794] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 07/30/2018] [Indexed: 12/17/2022] Open
Abstract
Hypoxic zones are common features of metastatic tumors. Due to inactivation of the von Hippel-Lindau gene (VHL), renal cell carcinomas (RCC) show constitutive stabilization of the alpha subunit of the hypoxia-inducible factor (HIF). Thus, RCC represents a model of chronic hypoxia. Development of the lymphatic network is dependent on vascular endothelial growth factor C (VEGFC) and lies at the front line of metastatic spreading. Here, we addressed the role of VEGFC in RCC aggressiveness and the regulation of its expression in hypoxia. Methods: Transcriptional and post transcriptional regulation of VEGFC expression was evaluated by qPCR and with reporter genes. The involvement of HIF was evaluated using a siRNA approach. Experimental RCC were performed with immuno-competent/deficient mice using human and mouse cells knocked-out for the VEGFC gene by a CRISPR/Cas9 method. The VEGFC axis was analyzed with an online available data base (TCGA) and using an independent cohort of patients. Results: Hypoxia induced VEGFC protein expression but down-regulated VEGFC gene transcription and mRNA stability. Increased proliferation, migration, over-activation of the AKT signaling pathway and enhanced expression of mesenchymal markers characterized VEGFC-/- cells. VEGFC-/- cells did not form tumors in immuno-deficient mice but developed aggressive tumors in immuno-competent mice. These tumors showed down-regulation of markers of activated lymphocytes and M1 macrophages, and up-regulation of M2 macrophages markers and programmed death ligand 1 (PDL1). Over-expression of lymphangiogenic genes including VEGFC was linked to increased disease-free and overall survival in patients with non-metastatic tumors, whereas its over-expression correlated with decreased progression-free and overall survival of metastatic patients. Conclusion: Our study revisited the admitted dogma linking VEGFC to tumor aggressiveness. We conclude that targeting VEGFC for therapy must be considered with caution.
Collapse
|
3178
|
A Critical Insight into the Clinical Translation of PD-1/PD-L1 Blockade Therapy in Clear Cell Renal Cell Carcinoma. Curr Urol Rep 2019; 20:1. [DOI: 10.1007/s11934-019-0866-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
3179
|
Patient-reported outcomes of patients with advanced renal cell carcinoma treated with nivolumab plus ipilimumab versus sunitinib (CheckMate 214): a randomised, phase 3 trial. Lancet Oncol 2019; 20:297-310. [PMID: 30658932 DOI: 10.1016/s1470-2045(18)30778-2] [Citation(s) in RCA: 193] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 10/09/2018] [Accepted: 10/11/2018] [Indexed: 12/19/2022]
Abstract
BACKGROUND In the ongoing phase 3, CheckMate 214 trial, nivolumab plus ipilimumab improved overall survival compared with sunitinib in patients with intermediate or poor risk, previously untreated, advanced renal cell carcinoma. We aimed to assess whether health-related quality of life (HRQoL) could be used to further describe the benefit-risk profile of nivolumab plus ipilimumab versus sunitinib. METHODS In the phase 3, randomised, controlled, CheckMate 214 trial, patients aged 18 years and older with previously untreated, advanced or metastatic renal cell carcinoma with a clear-cell component were recruited from 175 hospitals and cancer centres in 28 countries. Patients were categorised by risk status into favourable, intermediate, and poor risk subgroups and randomly assigned (1:1) to open-label nivolumab 3 mg/kg plus ipilimumab 1 mg/kg every 3 weeks for four doses followed by nivolumab 3 mg/kg every 2 weeks, or sunitinib 50 mg/day for 4 weeks of each 6-week cycle. Randomisation was done with a block size of four and stratified by risk status and geographical region. Patient-reported outcomes (PROs) were assessed using the Functional Assessment of Cancer Therapy Kidney Symptom Index-19 (FKSI-19), Functional Assessment of Cancer Therapy-General (FACT-G), and EuroQol five dimensional three level (EQ-5D-3L) instruments. The coprimary endpoints of the trial, reported previously, were overall survival, progression-free survival, and the proportion of patients who had an objective response in those categorised as at intermediate or poor risk. PROs in all randomised participants were assessed as an exploratory endpoint; here we report this exploratory endpoint. This study is registered with ClinicalTrials.gov, number NCT02231749, and is ongoing but is now closed to recruitment. FINDINGS Between Oct 16, 2014, and Feb 23, 2016, of 1390 patients screened, 1096 (79%) were randomly assigned to treatment, of whom 847 (77%) were at intermediate or poor risk and randomly assigned to nivolumab plus ipilimumab (n=425) or sunitinib (n=422). Median follow-up was 25·2 months (IQR 23·0-27·4). PROs were more favourable with nivolumab plus ipilimumab than sunitinib throughout the first 103 weeks after baseline, with mean change from baseline at week 103 for FKSI-19 total score being 4·00 (95% CI 1·91 to 6·09) for nivolumab plus ipilimumab versus -3·14 (-6·03 to -0·25) for sunitinib (p<0·0001), and for FACT-G total score being 4·77 (1·73 to 7·82) for nivolumab plus ipilimumab versus -4·32 (-8·54 to -0·11) for sunitinib (p=0·0005). Significant differences were also seen for four of five FKSI-19 domains (disease-related symptoms, physical disease-related symptoms, treatment side-effects, and functional wellbeing) and FACT-G physical and functional wellbeing domains. However, there was no significant difference between the treatment groups at week 103 in EQ-5D-3L visual analogue rating scale (VAS) scores, with mean change from baseline to week 103 of 10·07 (95% CI 4·35 to 15·80) for nivolumab plus ipilimumab and 6·40 (-1·36 to 14·16) for sunitinib (p=0·45). Compared with sunitinib, nivolumab plus ipilimumab reduced risk of deterioration in FKSI-19 total score (hazard ratio [HR] 0·54; 95% CI 0·46-0·63), FACT-G total score (0·63, 0·52-0·75), and EQ-5D-3L VAS score (HR 0·75, 95% CI 0·63-0·89) and UK utility scores (0·67, 0·57-0·80). INTERPRETATION Nivolumab plus ipilimumab leads to fewer symptoms and better HRQoL than sunitinib in patients at intermediate or poor risk with advanced renal cell carcinoma. These results suggest that the superior efficacy of nivolumab plus ipilimumab over sunitinib comes with the additional benefit of improved HRQoL. FUNDING Bristol-Myers Squibb and ONO Pharmaceutical.
Collapse
|
3180
|
Mery B, Jones S, Vallard A, Rowinski E, Guillot A, Magné N. Cancer du rein métastatique : recommandations et perspectives en 1re ligne. Bull Cancer 2019; 105 Suppl 3:S235-S241. [PMID: 30595152 DOI: 10.1016/s0007-4551(18)30378-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
METASTATIC RENAL CELL CARCINOMA WHICH TREATMENTS IN FIRST-LINE SETTING?: The treatment of metastatic kidney cancer has radically changed during the past decade, notably with the development of tyrosin kinase inhibitors (TKI) and the rise of immunotherapy. Kidney cancer, especially clear cell renal cell carcinoma (CCRC) which regroups 80% of cases, is associated with increased angiogenesis and VEGF (vascular endothelial growth factor) dependent signaling pathways. Targeted therapies have therefore modified therapeutical strategies through direct inhibition of VEGF on its receptor or inhibition of the PI3K/AKT/mTOR pathway. Consequently, new anti-angiogenic molecules are now available as first line treatment and are to be prioritized depending on tumoral histology and prognostic groups. These new molecules have allowed increased patient survival. Immunotherapy is again currently transforming our first line therapeutical approach of metastatic kidney cancer with numerous ongoing therapeutical trials including combination of targeted therapies with immune checkpoint inhibitors or association of various immunotherapies. Beyond these major first line changes, difficulties still remain in the therapeutical sequence which is crucial in the care of these patients. This report aims to underline first line therapeutical recommendations in metastatic kidney cancer and expose results of recent assays.
Collapse
Affiliation(s)
- Benoîte Mery
- Département d'oncologie médicale, Institut de Cancérologie Lucien Neuwirth, Saint Priest en Jarez, France
| | - Samuel Jones
- Centre Hospitalier Universitaire Vaudois, Lausanne, Suisse
| | - Alexis Vallard
- Département de radiothérapie, Institut de Cancérologie Lucien Neuwirth, Saint Priest en Jarez, France
| | - Elise Rowinski
- Département d'oncologie médicale, Institut de Cancérologie Lucien Neuwirth, Saint Priest en Jarez, France
| | - Aline Guillot
- Département d'oncologie médicale, Institut de Cancérologie Lucien Neuwirth, Saint Priest en Jarez, France
| | - Nicolas Magné
- Département de radiothérapie, Institut de Cancérologie Lucien Neuwirth, Saint Priest en Jarez, France.
| |
Collapse
|
3181
|
Amin A, Hammers H. The Evolving Landscape of Immunotherapy-Based Combinations for Frontline Treatment of Advanced Renal Cell Carcinoma. Front Immunol 2019; 9:3120. [PMID: 30687324 PMCID: PMC6335326 DOI: 10.3389/fimmu.2018.03120] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 12/18/2018] [Indexed: 12/25/2022] Open
Abstract
Insights into the biology of advanced renal cell carcinoma (aRCC) and the development of agents targeting the vascular endothelial growth factor (VEGF) pathway have positively impacted the outcomes for patients with aRCC. With the recent approval of the dual immune checkpoint inhibitors (ICIs), nivolumab and ipilimumab, by the U.S. Food and Drug Administration (USFDA), and the European Medicines Agency (EMA), the era of VEGF monotherapy for untreated aRCC appears to be coming to an end for patients with access to the combination therapy. The frontline treatment options for renal cell carcinoma are evolving rapidly and will lead to the approval of other combination immunotherapies-especially those with VEGF inhibitors. Here we review the clinical data for dual immune checkpoint inhibition with nivolumab plus ipilimumab as well as the emerging data for ICI plus VEGF inhibitor combinations and discuss the challenges these will pose for the clinical practitioner.
Collapse
Affiliation(s)
- Asim Amin
- Levine Cancer Institute, Charlotte, NC, United States
- Atrium Healthcare System, Charlotte, NC, United States
| | - Hans Hammers
- University of Texas Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
3182
|
Chen RQ, Liu F, Qiu XY, Chen XQ. The Prognostic and Therapeutic Value of PD-L1 in Glioma. Front Pharmacol 2019; 9:1503. [PMID: 30687086 PMCID: PMC6333638 DOI: 10.3389/fphar.2018.01503] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Accepted: 12/10/2018] [Indexed: 12/19/2022] Open
Abstract
Glioma is the most common type of primary brain tumors. After standard treatment regimen (surgical section, radiotherapy and chemotherapy), the average survival time remains merely around 14 months for glioblastoma (grade IV glioma). Recent immune therapy targeting to the immune inhibitory checkpoint axis, i.e., programmed cell death protein 1 (PD-1) and its ligand PD-L1 (i.e., CD274 or B7-H1), has achieved breakthrough in many cancers but still not in glioma. PD-L1 is considered a major prognostic biomarker for immune therapy in many cancers, with anti-PD-1 or anti-PD-L1 antibodies being used. However, the expression and subcellular distribution of PD-L1 in glioma cells exhibits great variance in different studies, severely impairing PD-L1's value as therapeutic and prognostic biomarker in glioma. The role of PD-L1 in modulating immune therapy is complicated. In addition, endogenous PD-L1 plays tumorigenic roles in glioma development. In this review, we summarize PD-L1 mRNA expression and protein levels detected by using different methods and antibodies in human glioma tissues in all literatures, and we evaluate the prognostic value of PD-L1 in glioma. We also summarize the relationships between PD-L1 and immune cell infiltration in glioma. The mechanisms regulating PD-L1 expression and the oncogenic roles of endogenous PD-L1 are discussed. Further, the therapeutic results of using anti-PD-1/PD-L1 antibodies or PD-L1 knockdown are summarized and evaluated. In summary, current results support that PD-L1 is not only a prognostic biomarker of immune therapy, but also a potential therapeutic target for glioma.
Collapse
Affiliation(s)
- Ruo Qiao Chen
- School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Feng Liu
- School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xin Yao Qiu
- School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiao Qian Chen
- School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
3183
|
Tsimafeyeu I, Borisov P, Abdelgafur A, Leonenkov R, Novikova O, Guseva I, Demchenkova M, Mikhailova N, Semenov A, Yurmazov Z, Sivunova I, Ramazanova M, Gamayunov S, Kosov D, Bratslavsky G. Phase 2 Multicenter Single-Arm Study of Second-Line Axitinib in Favorable Risk Patients with Metastatic Renal Cell Carcinoma: FavorAx. Target Oncol 2019; 14:33-38. [PMID: 30607698 DOI: 10.1007/s11523-018-0613-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
BACKGROUND Targeted therapy with axitinib resulted in a greater objective response rate and prolonged progression-free survival (PFS) compared to sorafenib in patients with previously treated metastatic renal cell carcinoma (mRCC) in the phase 3 AXIS study, where 75% of patients had intermediate and poor International Metastatic Renal Cell Carcinoma Database Consortium (IMDC) risk. OBJECTIVE In this phase 2 study (FavorAx), we assessed the activity of axitinib in mRCC patients with a favorable risk and history of prior vascular endothelial growth factor receptor (VEGFR)-directed therapy. PATIENTS AND METHODS Patients were required to have clear-cell mRCC, favorable risk according to IMDC criteria, and to have received first-line treatment with sunitinib or pazopanib. Prior treatment with other agents was not permitted. The primary endpoint of the study was 5 months PFS. Additional endpoints included response rate, safety, PFS, and overall survival (OS). RESULTS A total of 21 patients were enrolled, 62% of whom were male. The mean age was 60 years. Eleven (52%) patients had two or more metastatic sites. 67% and 33% of patients received first-line sunitinib or pazopanib, respectively, with a median PFS of 17 months [95% confidence interval (CI), 14-20]. After a median follow-up of 25 months, the median PFS was 19 months (95% CI, 15-23). The current study did achieve its primary endpoint based on the 5-month PFS of 100%. The median OS was not yet reached. The 18 months OS rate was 85.7%. The objective response rate was 33% and one patient achieved a complete response. Seven patients had dose escalation of axitinib and four patients had dose reduction. Grade 3 adverse events were observed in 19% of cases. There was no discontinuation of therapy due to toxicity. CONCLUSIONS The encouraging PFS and favorable safety profile observed in the FavorAx study support the administration of axitinib in mRCC patients with favorable IMDC risk and a history of prior sunitinib or pazopanib.
Collapse
Affiliation(s)
- Ilya Tsimafeyeu
- Kidney Cancer Research Bureau, Mayakovskogo pereulok, 2, 109147, Moscow, Russia.
| | - Pavel Borisov
- City Clinical Oncology Center, St. Petersburg, Russia
| | | | | | - Olga Novikova
- Khabarovsk Regional Cancer Center, Khabarovsk, Russia
| | | | | | | | | | | | - Irina Sivunova
- Kamchatka Regional Cancer Center, Petropavlovsk-Kamchatsky, Russia
| | | | | | - Dmitry Kosov
- Aston Health Contract Research Organization, Moscow, Russia
| | | |
Collapse
|
3184
|
Adjuvant Tyrosine Kinase Inhibitors in Treatment of Renal Cell Carcinoma: A Meta-Analysis of Available Clinical Trials. Clin Genitourin Cancer 2019; 17:e339-e344. [PMID: 30704796 DOI: 10.1016/j.clgc.2018.12.011] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 12/27/2018] [Accepted: 12/30/2018] [Indexed: 11/23/2022]
Abstract
Although tyrosine kinase inhibitors (TKIs) are widely used in the metastatic setting, they have shown more limited effectiveness in the adjuvant setting. Despite multiple clinical trials, there has been no improvement in overall survival (OS) with the use of these agents as adjuvant therapy, and conflicting data on disease-free survival (DFS). We carried out a meta-analysis of available phase III randomized clinical trials on adjuvant TKIs in clear-cell renal cell carcinoma (RCC). Primary end points of our study were the effect of adjuvant TKIs on OS and DFS in the overall population. Furthermore, we investigated if use of adjuvant TKIs resulted in improved DFS among patients with different risk of tumor relapse. Higher-risk patients were patients with 1 or more of the following features: positive nodes (N+), T4 tumors and T3 tumors, with higher Fuhrman grades (3-4). We adopted Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines to carry out our study. In the overall population, the pooled hazard ratio (HR) of OS and DFS was 0.89 (95% confidence interval [CI], 0.76-1.04) and 0.84 (95% CI, 0.76-0.93), respectively. In the low- and high-risk populations, the pooled DFS HR was 0.98 (95% CI, 0.82-1.17) and 0.85 (95% CI, 0.75-0.97), respectively. Adjuvant use of TKIs do not appear to provide a statistically significant OS benefit. However, a benefit in DFS has been observed in overall and high-risk populations, suggesting that better selection of patients might be important for the evaluation of adjuvant therapies in RCC.
Collapse
|
3185
|
External validation of the systemic immune-inflammation index as a prognostic factor in metastatic renal cell carcinoma and its implementation within the international metastatic renal cell carcinoma database consortium model. Int J Clin Oncol 2019; 24:526-532. [PMID: 30604160 DOI: 10.1007/s10147-018-01390-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 12/26/2018] [Indexed: 12/31/2022]
Abstract
BACKGROUND We conducted a study to validate the influence of the systemic immune-inflammation index (SII) on overall survival (OS) in patients with metastatic renal cell carcinoma (mRCC) and to verify whether the implementation of the SII in place of neutrophil and platelet counts within the International Metastatic Renal Cell Carcinoma Consortium (IMDC) model might increase its prognostic accuracy. PATIENTS AND METHODS We retrospectively analyzed consecutive patients with mRCC, who were treated with first-line tyrosine kinase inhibitors from 2008 to 2016 in two major oncology centres in Poland. We stratified patients into low SII (< 730) and high SII (≥ 730) groups according to a recent literature report. We used multivariable Cox proportional hazards regressions (CPHRs) to assess the impact of the SII on OS and concordance, global 'goodness-of-fit', calibration and reclassification measures to quantify a potential prognostic benefit from the modification of the IMDC model. RESULTS Overall, 502 patients (294 with low and 208 with high SII) were included. Median OS was 36.7 months [95% confidence interval (CI) 30.4-41.5 months] and 17.0 months (95% CI 12.5-19.6 months) in the low and high SII groups, respectively. The SII status was significant in CPHRs with the hazard ratio ranging from 1.38 to 1.68. All prognostic accuracy measures favored the SII-modified-IMDC model over the original IMDC model. CONCLUSIONS Using an external dataset, we showed that high SII was an independent factor for poor OS. The addition of the SII to the IMDC model in place of neutrophil and platelet counts increased the model's prognostic performance.
Collapse
|
3186
|
Schulster M. Bladder Cancer Academy 2019 Selected Summaries. Rev Urol 2019; 21:23-28. [PMID: 31239827 PMCID: PMC6585182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Affiliation(s)
- Michael Schulster
- Department of Urology, NYU Langone Health Medical Center New York, NY
| |
Collapse
|
3187
|
Guida A, Albiges L. Traitement des métastases osseuses du cancer du rein. Bull Cancer 2019; 105 Suppl 3:S268-S279. [PMID: 30595156 DOI: 10.1016/s0007-4551(18)30382-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
TREATING PATIENTS WITH BONE METASTASES FROM RENAL CELL CARCINOMA Bone metastases (BMs) are common and cause morbidity in cancer patients. One third of metastatic renal cancer (mRCC) patients present metastatic disease to the bone. BMs cause severe complications such as fracture, spinal cord compression and pain requiring surgery or radiotherapy. Hypercalcaemia is a common feature in mRCC as well as an established poor prognosis factor. BMs impact negatively on prognosis and affect quality of life. Correct management of BMs from RCC requires a multimodal evaluation to optimize care and quality of life. In this review, we discuss current evidences on the role of systemic treatments in BMs management, bone-targeting agents benefits in skeletal-related events prevention and local therapeutic approaches to BM in mRCC. Define prognosis of systemic disease and identify the main goal of treatment are crucial for the selection of the best strategy.
Collapse
Affiliation(s)
| | - Laurence Albiges
- Département d'oncologie médicale, Gustave Roussy, Villejuif, France..
| |
Collapse
|
3188
|
Systematic Review of the Role of Cytoreductive Nephrectomy in the Targeted Therapy Era and Beyond: An Individualized Approach to Metastatic Renal Cell Carcinoma. Eur Urol 2019; 75:111-128. [DOI: 10.1016/j.eururo.2018.09.016] [Citation(s) in RCA: 106] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 09/10/2018] [Indexed: 01/02/2023]
|
3189
|
Fares CM, Van Allen EM, Drake CG, Allison JP, Hu-Lieskovan S. Mechanisms of Resistance to Immune Checkpoint Blockade: Why Does Checkpoint Inhibitor Immunotherapy Not Work for All Patients? Am Soc Clin Oncol Educ Book 2019; 39:147-164. [PMID: 31099674 DOI: 10.1200/edbk_240837] [Citation(s) in RCA: 468] [Impact Index Per Article: 78.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/14/2023]
Abstract
The emergence of immune checkpoint blockade therapies over the last decade has transformed cancer treatment in a wide range of tumor types. Unprecedented and durable clinical responses in difficult-to-treat cancer histologies have been observed. However, despite these promising long-term responses, the majority of patients fail to respond to immune checkpoint blockade, demonstrating primary resistance. Additionally, many of those who initially respond to treatment eventually experience relapse secondary to acquired resistance. Both primary and acquired resistance are a result of complex and constantly evolving interactions between cancer cells and the immune system. Many mechanisms of resistance have been characterized to date, and more continue to be uncovered. By elucidating and targeting mechanisms of resistance, treatments can be tailored to improve clinical outcomes. This review will discuss the landscape of immune checkpoint blockade response data, different resistance mechanisms, and potential therapeutic strategies to overcome resistance.
Collapse
Affiliation(s)
- Charlene M Fares
- 1 Department of Medicine, Division of Hematology/Oncology, University of California, Los Angeles, Los Angeles, CA
| | | | - Charles G Drake
- 3 Department of Medicine, Division of Hematology/Oncology, Columbia University Medical Center, New York, NY
| | - James P Allison
- 4 Department of Immunology, Division of Basic Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Siwen Hu-Lieskovan
- 5 Division of Hematology and Oncology, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| |
Collapse
|
3190
|
Vano YA, Simonaggio A, Thibault C, Oudard S. Immunothérapie des cancers du rein. Bull Cancer 2019; 105 Suppl 1:S24-S34. [PMID: 30595195 DOI: 10.1016/s0007-4551(18)30387-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
IMMUNOTHERAPY FOR RENAL CELL CARCINOMA Clear cell kidney cancer is a tumour type whose development and progression is driven by the HIF/VEGF angiogenesis pathway. Anti-angiogenic (AA) agents, particularly anti-VEGFR tyrosine kinase (TKI) inhibitors, have profoundly modified the prognosis of patients in the metastatic setting (mRCC) since their registration in 2006. At the same time, mTOR inhibitors have also brought significant benefit to patients. More recently, treatments restoring adaptive anti-tumor immunity, anti-program death 1 (anti-PD-1) checkpoint inhibitors (ICP), have in turn revolutionized the management of patients with mRCC. The multi-tumor efficacy of these ICPs proves the crucial role of anti-tumor immunity in tumor development and progression in a number of tumors including clear cell kidney tumours (ccRCC). The tumor immune microenvironment (TME) of ccRCC is known to be highly immunosuppressive. Thus ccRCCs are characterized by a strong infiltration of CD8+ T lymphocytes, frequently expressing immunological checkpoint molecules on their surface, giving them a poor prognostic feature. These characteristics partly explain the effectiveness of ICP in ccRCC and constitute a strong rationale for their further development, either at an earlier stage or in combination, particularly with AA or TKI. In this review are compiled the main clinical results of immunological checkpoint molecules alone or in combination in 1stline or after TKI failure.
Collapse
Affiliation(s)
- Yann-Alexandre Vano
- Service d'oncologie médicale, Hôpital européen Georges-Pompidou, Paris, France; Inserm, UMRS 1138, Centre de recherche des cordeliers, cancer et immunité antitumorale, université Paris-Descartes, Sorbonne-Paris-Cité, F-75006, Paris, France.
| | - Audrey Simonaggio
- Service d'oncologie médicale, Hôpital européen Georges-Pompidou, Paris, France; Inserm, UMRS 1138, Centre de recherche des cordeliers, cancer et immunité antitumorale, université Paris-Descartes, Sorbonne-Paris-Cité, F-75006, Paris, France
| | - Constance Thibault
- Service d'oncologie médicale, Hôpital européen Georges-Pompidou, Paris, France; Inserm, UMRS 1138, Centre de recherche des cordeliers, cancer et immunité antitumorale, université Paris-Descartes, Sorbonne-Paris-Cité, F-75006, Paris, France
| | - Stéphane Oudard
- Service d'oncologie médicale, Hôpital européen Georges-Pompidou, Paris, France; Immunothérapies et antiangiogéniques en oncologie, équipe 10 PARCC, INSERM U970, Sorbonne-Paris-Cité, Paris, France
| |
Collapse
|
3191
|
Sharma A, Suleyman N, Jones O, Vasdev N. Immunotherapy in Urological Tumors. Rev Urol 2019; 21:15-20. [PMID: 31239825 PMCID: PMC6585187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
The past decade has seen significant improvement in our understanding of tumor biological features, which has led to use of anti-programmed-death 1 (PD-1) and anti-PD-ligand-1 (PD-L1) agents and cytotoxic T lymphocytes antigen 4 (CTLA-4) inhibitors in a multitude of cancers. These immunotherapeutic agents have shown activity in melanoma, lung, head and neck, colorectal, urological, and other cancers. This article details the use of immunotherapy agents in urothelial, renal, prostate, and testicular tumors.
Collapse
Affiliation(s)
- Anand Sharma
- Department of Oncology, Mount Vernon Cancer Centre Northwood, UK
| | - Narin Suleyman
- Hertfordshire and Bedfordshire Urological Cancer Centre and Department of Urology and Surgery, Lister Hospital, East and North Herts NHS Trust Stevenage, UK
| | - Oliver Jones
- Hertfordshire and Bedfordshire Urological Cancer Centre and Department of Urology and Surgery, Lister Hospital, East and North Herts NHS Trust Stevenage, UK
| | | |
Collapse
|
3192
|
Bersanelli M, Gnetti L, Varotti E, Ampollini L, Carbognani P, Leonardi F, Rusca M, Campanini N, Ziglioli F, Dadomo CI, Pilato FP, Cortellini A, Rapacchi E, Caruso G, Silini EM, Maestroni U, Buti S. Immune context characterization and heterogeneity in primary tumors and pulmonary metastases from renal cell carcinoma. Immunotherapy 2019; 11:21-35. [PMID: 30702014 DOI: 10.2217/imt-2018-0097] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Accepted: 10/11/2018] [Indexed: 12/25/2022] Open
Abstract
AIM The knowledge of the immune context of renal cell carcinoma (RCC) is useful to predict benefit from immunotherapy. We retrospectively characterized the immune context of RCC patients underwent primary nephrectomy and pulmonary metastasectomy. MATERIALS & METHODS Intratumoral infiltrating lymphocytes and peritumoral renal infiltrating lymphocytes, lymphocyte subpopulations (CD4+, CD8+), PD-1, PD-L1 were explored in paired samples of primary RCC (T) and respective pulmonary metastases (M). RESULTS The immune variables demonstrated intralesional and intratumoral heterogeneity. Intralesional lymphocyte heterogeneity reached 76% of cases in T, 28% in M. The heterogeneity rate for PD-L1 expression was from 44% (T) to 56% (M); it correlated with better survival. CONCLUSION The immune context of RCC is highly variable both within a given tumor and among primary and metastases.
Collapse
Affiliation(s)
- Melissa Bersanelli
- Department of Medical Oncology, University Hospital of Parma, Parma, 43126, Italy
| | - Letizia Gnetti
- Department of Pathological Anatomy & Histology, University Hospital of Parma, Parma, 43126, Italy
| | - Elena Varotti
- Department of Pathological Anatomy & Histology, University Hospital of Parma, Parma, 43126, Italy
| | - Luca Ampollini
- Department of Thoracic Surgery, University Hospital of Parma, Parma, 43126, Italy
| | - Paolo Carbognani
- Department of Thoracic Surgery, University Hospital of Parma, Parma, 43126, Italy
| | - Francesco Leonardi
- Department of Medical Oncology, University Hospital of Parma, Parma, 43126, Italy
| | - Michele Rusca
- Department of Thoracic Surgery, University Hospital of Parma, Parma, 43126, Italy
| | - Nicoletta Campanini
- Department of Pathological Anatomy & Histology, University Hospital of Parma, Parma, 43126, Italy
| | - Francesco Ziglioli
- Department of Urology, University Hospital of Parma, Parma, 43126, Italy
| | - Clara I Dadomo
- Department of Pathological Anatomy & Histology, University Hospital of Parma, Parma, 43126, Italy
| | - Francesco P Pilato
- Department of Pathological Anatomy & Histology, University Hospital of Parma, Parma, 43126, Italy
| | - Alessio Cortellini
- Department of Biotechnological & Applied Clinical Sciences, Medical Oncology, St Salvatore Hospital, University of L'Aquila, L'Aquila, 67100, Italy
| | - Elena Rapacchi
- Department of Medical Oncology, University Hospital of Parma, Parma, 43126, Italy
| | - Giuseppe Caruso
- Department of Medical Oncology, University Hospital of Parma, Parma, 43126, Italy
| | - Enrico M Silini
- Department of Pathological Anatomy & Histology, University Hospital of Parma, Parma, 43126, Italy
| | - Umberto Maestroni
- Department of Urology, University Hospital of Parma, Parma, 43126, Italy
| | - Sebastiano Buti
- Department of Medical Oncology, University Hospital of Parma, Parma, 43126, Italy
| |
Collapse
|
3193
|
Grivas P, Drakaki A, Friedlander TW, Sonpavde G. Conceptual Framework for Therapeutic Development Beyond Anti-PD-1/PD-L1 in Urothelial Cancer. Am Soc Clin Oncol Educ Book 2019; 39:284-300. [PMID: 31099684 DOI: 10.1200/edbk_237449] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Platinum-based chemotherapy has been the standard of care in advanced urothelial cancer, but long-term outcomes have remained poor. Immune checkpoint inhibitors, with their favorable toxicity profiles and noteworthy efficacy, have steered a new era in advanced urothelial cancer, with five agents targeting the PD-1/PD-L1 pathway approved by the U.S. Food and Drug Administration (FDA). However, most patients do not achieve response, whereas immunotherapy-related adverse events may cause morbidity, increased health care use, and-rarely-mortality. Therefore, there is an urgent need for additional therapeutic modalities across the disease spectrum. A plethora of clinical trials are ongoing in various disease settings, including chemotherapy regimens, radiotherapy, antibody-drug conjugates, agents targeting additional immune checkpoint pathways, vaccine, cytokines, adoptive cell therapies, as well as targeted and anti-angiogenic agents. Two agents, enfortumab vedotin and erdafitinib, have breakthrough designation by the FDA but are not approved yet (at the time of this paper's preparation). Novel combinations with various treatment modalities and optimal sequencing of active therapies are being investigated in prospective clinical trials. Evaluation of new treatments has met with substantial challenges for many reasons, for example, molecular heterogeneity, clonal evolution, and genomic instability. In the era of precision molecular medicine, and because patients do not respond uniformly to current therapies, there is a growing need for identification and validation of biomarkers that can accurately predict treatment response and assist in patient selection. Here, we review current updates and future directions of experimental therapeutics in urothelial cancer, including examples (but not an exhaustive list) of ongoing clinical trials.
Collapse
Affiliation(s)
- Petros Grivas
- 1 From the University of Washington, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Alexandra Drakaki
- 2 David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA
| | - Terence W Friedlander
- 3 Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA
| | - Guru Sonpavde
- 4 Dana Farber Cancer Institute, Harvard Medical School, Boston, MA
| |
Collapse
|
3194
|
Dahl O, Brydøy M. The pioneers behind immune checkpoint blockers awarded the Nobel Prize in physiology or medicine 2018. Acta Oncol 2019; 58:1-8. [PMID: 30698061 DOI: 10.1080/0284186x.2018.1555375] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Olav Dahl
- Department of Clinical Science Faculty of Medicine, University of Bergen, Bergen, Norway
- Department of Oncology and Medical Physics, Haukeland University Hospital, Bergen, Norway
| | - Marianne Brydøy
- Department of Oncology and Medical Physics, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
3195
|
Kondisetty G, Borkar PV, Kondisetty S, Thomas A. Retrospective review of experience with sarcomatoid renal cell carcinoma: Multimodality treatment remains an unmet goal. Urol Ann 2019; 11:385-388. [PMID: 31649458 PMCID: PMC6798302 DOI: 10.4103/ua.ua_106_18] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Background: Sarcomatoid change in Renal cell carcinoma(RCC) is associated with adverse outcomes with median survival of 6 months. Settings and Design: This is a retrospective study of patients diagnosed of sarcomatoid RCC(sRCC) between 2007 and 2013 which were followed up till 2017. Methods and Material: Patients (n=22) were grouped based on whether they received additional chemotherapy following nephrectomy. Two groups were followed up until 2017 and overall survival was record. Overall survival curves were estimated by Kaplan-Meier method and compared using Log Rank (Mantel-Cox) test between two groups. Statistical analysis used: Kaplan-Meier method and Log Rank (Mantel-Cox) test. Results: The patients who had chemotherapy had 13.4 cm of mean tumour size with a mean survival of 20.4 ± 8.3 months. The patients who did not undergo chemotherapy had mean tumour size of 11.7 cm with a mean survival of 21 ± 5.9 months. There was no much statistical difference between the two groups in OS with P value = 0.99. Conclusion: The current adjuvant chemotherapy used in sRCC patients who develop metastasis gives no survival advantage.
Collapse
Affiliation(s)
- Gowrinath Kondisetty
- Department of General Surgery, Medicity Institute of Medical Sciences, Medchal, Telangana, India
| | - Pallavi Vijay Borkar
- Department of Pathology, Amrita Institute of Medical Sciences, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| | - Sandeep Kondisetty
- Department of Urology, Amrita Institute of Medical Sciences, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| | - Appu Thomas
- Department of Urology, Amrita Institute of Medical Sciences, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| |
Collapse
|
3196
|
Psutka SP, Chang SL, Cahn D, Uzzo RG, McGregor BA. Reassessing the Role of Cytoreductive Nephrectomy for Metastatic Renal Cell Carcinoma in 2019. Am Soc Clin Oncol Educ Book 2019; 39:276-283. [PMID: 31099657 DOI: 10.1200/edbk_237453] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Cytoreductive nephrectomy (CRN) has long been considered a standard of care in the management of mRCC. This is largely based on randomized trials in the era of interferon (IFN) that demonstrate an improvement in overall survival (OS). With the advent of targeted therapies, the role of CRN has been questioned and multiple retrospective analyses have shown a potential benefit, particularly in intermediate-risk disease. Two long-awaited prospective trials have been published in the past year that explore the role of CRN. The CARMENA trial randomly assigned patients to therapy with sunitinib with or without CRN, showing noninferiority of sunitinib alone versus sunitinib plus CRN with a median OS of 18.4 months versus 13.9 months, respectively (hazard ratio [HR] for mortality, 0.89; 95% CI, 0.71-1.1). The SURTIME trial randomly assigned patients to immediate CRN followed by sunitinib versus a deferred CRN after three cycles of sunitinib. Analysis is limited by early termination as a result of low accrual. Although there was no difference in progression-free survival (PFS), median OS was significantly improved among patients in the deferred CRN arm (HR, 0.57; 95% CI, 0.34-0.95; p = .032). Early systemic therapy is paramount, but there are patients who may derive benefit by incorporating the removal of the primary tumor in their multimodal therapy, perhaps in a deferred setting. As systemic treatment paradigms shift and immunotherapy again moves to the frontline setting with the potential for novel therapeutic approaches, the role of CRN will continue to evolve with the potential to offer surgical interventions with minimal, if any, delay in systemic treatment.
Collapse
Affiliation(s)
- Sarah P Psutka
- 1 Department of Urology, University of Washington, Seattle, WA
| | - Steven L Chang
- 2 Brigham and Women's Hospital, Boston, MA
- 3 Dana-Farber Cancer Institute, Boston, MA
| | - David Cahn
- 4 Fox Chase Cancer Center, Philadelphia, PA
| | | | - Bradley A McGregor
- 3 Dana-Farber Cancer Institute, Boston, MA
- 5 Harvard Medical School, Boston, MA
| |
Collapse
|
3197
|
Prabhash K, Patil V, Noronha V, Joshi A, Abhyankar A, Menon N, Banavali S, Gupta S. Low doses in immunotherapy: Are they effective? CANCER RESEARCH, STATISTICS, AND TREATMENT 2019. [DOI: 10.4103/crst.crst_29_19] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
3198
|
Radiological Response and Neutrophil-to-Lymphocyte Ratio as Predictive Factors for Progression-Free and Overall Survival in Metastatic Renal Cell Carcinoma Patients Treated with Sunitinib. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1153:31-45. [PMID: 30903615 DOI: 10.1007/5584_2019_352] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Renal cell carcinoma (RCC) represents 2-3% of all malignancies. Most RCC-related deaths are caused by metastases of the disease. Studies suggest that inflammation-related parameters are of prognostic significance in metastatic renal cell carcinoma (mRCC) patients. Neutrophilia and thrombocytosis are markers of systemic inflammation that accompanies cancer, while lymphopenia is related to dysfunctions of the immune system. Neutrophil-to-lymphocyte ratio (NLR) and platelet-to-lymphocyte ratio (PLR) thus seem particularly interesting from a clinical perspective. The goal of this study was to determine if the response to therapy, consisting of reductions in radiologically assessed tumor burden and in inflammation-related parameters after 12 weeks of treatment with sunitinib, has a predictive value for outcome. One hundred thirty-one mRCC patients treated with the first-line sunitinib were evaluated. Inflammation-related parameters and radiologic response were correlated with treatment outcomes, progression-free, and overall survival. We found that the longest median progression-free survival of 37 months (Q1; Q3-15; not reached) and overall survival of 40 months (Q1; Q3-26; not reached) were achieved by patients who had either partial or complete response according to RECIST 1.1 and NLR lower than 1.64. In conclusion, the study confirmed that both objective response and lower grade of inflammation during treatment are predictive of better outcomes in mRCC patients treated with sunitinib.
Collapse
|
3199
|
Rios A, Salazar GT, Zhang N, An Z. The 2018 Nobel Prize in Medicine for breakthroughs in targeting immune checkpoint inhibitors: a brief perspective. Antib Ther 2019; 2:40-43. [PMID: 33928220 PMCID: PMC7990146 DOI: 10.1093/abt/tbz003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 02/06/2019] [Accepted: 02/21/2019] [Indexed: 12/15/2022] Open
Affiliation(s)
- Adan Rios
- Division of Oncology, Department of Internal Medicine
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Georgina To'a Salazar
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Ningyan Zhang
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Zhiqiang An
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| |
Collapse
|
3200
|
Salabert L, Sionneau B, Cochin V, Ravaud A, Gross-Goupil M. Traitement focal et traitement systémique dans la prise en charge du cancer du rein métastatique : une question de complémentarité. Bull Cancer 2019; 105 Suppl 3:S221-S228. [PMID: 30595150 DOI: 10.1016/s0007-4551(18)30376-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
FOCAL TREATMENT AND SYSTEMIC THERAPY IN METASTATIC KIDNEY CANCER COMPLEMENTARY APPROACHES: To treat metastatic renal cell carcinoma, therapies used are with an oral intake, for the vast majority, and have many side effects that may compromise observance. Strategies of drug holiday have been studied and, in case of an indolent and oligometastatic tumor, studies have shown that active surveillance is possible to delay the introduction of systemic treatment, without compromising the patient survival. A multimodal approach combining systemic and focal treatments can be done with several objectives: to delay even more introduction of systemic treatment by focally treating metastases, to allow drug holiday after partial response to medical treatment by local control of persistent metastases, and to permit drug continuation even in case of dissociated response to systemic therapy, by focal treatment of metastasis(es) in progression. Technics that can be used for focal treatment are metastasectomy, radiofrequency ablation or cryotherapy, and stereotactic radiotherapy. In literature, studies that evaluated this approach are for almost retrospective studies, but they have reported interesting results in terms of local control and low morbidity. In the era of checkpoint's inhibitors, it seems important to make prospective collections of data to validate these practices. In any case, and because international recommendations about multimodal approach are poor, discussions between the different actors of the patient care are essential to find the most beneficial treatment for him.
Collapse
Affiliation(s)
- Laura Salabert
- Service oncologie médicale, centre hospitalo-universitaire Bordeaux, France
| | - Baptiste Sionneau
- Service oncologie médicale, centre hospitalo-universitaire Bordeaux, France
| | - Valérie Cochin
- Service oncologie médicale, centre hospitalo-universitaire Bordeaux, France
| | - Alain Ravaud
- Service oncologie médicale, centre hospitalo-universitaire Bordeaux, France; Université de Bordeaux, France
| | - Marine Gross-Goupil
- Service oncologie médicale, centre hospitalo-universitaire Bordeaux, France.
| |
Collapse
|