1
|
Samnani S, Sachedina F, Gupta M, Guo E, Navani V. Mechanisms and clinical implications in renal carcinoma resistance: narrative review of immune checkpoint inhibitors. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2023; 6:416-429. [PMID: 37457122 PMCID: PMC10344724 DOI: 10.20517/cdr.2023.02] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 05/25/2023] [Accepted: 06/13/2023] [Indexed: 07/18/2023]
Abstract
Clear cell renal cell carcinoma (ccRCC) is the most common histological subtype of renal cell carcinoma. The prognosis for patients with ccRCC has improved over recent years with the use of combination therapies with an anti-programmed death-1 (PD-1) backbone. This has enhanced the quality of life and life expectancy of patients with this disease. Unfortunately, not all patients benefit; eventually, most patients will develop resistance to therapy and progress. Recent molecular, biochemical, and immunological research has extensively researched anti-angiogenic and immune-based treatment resistance mechanisms. This analysis offers an overview of the principles underpinning the resistance pathways related to immune checkpoint inhibitors (ICIs). Additionally, novel approaches to overcome resistance that may be considered for the trial context are discussed.
Collapse
Affiliation(s)
- Sunil Samnani
- Department of Internal Medicine, The University of Calgary, Calgary T2N 1N4, Canada
| | - Faraz Sachedina
- Department of Internal Medicine, The University of Calgary, Calgary T2N 1N4, Canada
| | - Mehul Gupta
- Cumming School of Medicine, University of Calgary, Calgary T2N 4N1, Canada
| | - Edward Guo
- Cumming School of Medicine, University of Calgary, Calgary T2N 4N1, Canada
| | - Vishal Navani
- Department of Medical Oncology, Tom Baker Cancer Centre, Calgary T2N 4N2, Canada
| |
Collapse
|
2
|
Larroquette M, Lefort F, Heraudet L, Bernhard JC, Ravaud A, Domblides C, Gross-Goupil M. Therapeutic Management of Metastatic Clear Cell Renal Cell Carcinoma: A Revolution in Every Decade. Cancers (Basel) 2022; 14:6230. [PMID: 36551715 PMCID: PMC9777357 DOI: 10.3390/cancers14246230] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/13/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
Clear cell renal cell carcinoma (RCC) oncogenesis is mainly driven by VHL gene inactivation, leading to overexpression of vascular endothelial growth factor (VEGF). The use of tyrosine-kinase inhibitors (TKIs) directed against VEGF and its receptor (VEGFR) revolutionised the management of metastatic renal cancer in the 2000s. The more recent development of next-generation TKIs such as cabozantinib or lenvatinib has made it possible to bypass some of the mechanisms of resistance to first-generation anti-VEGFR TKIs. During the decade 2010-2020, the development of immune checkpoint blockade (ICB) therapies revolutionised the management of many solid cancers, including RCC, in first- and subsequent-line settings. Dual ICB or ICB plus anti-VEGFR TKI combinations are now the standard of care for patients with advanced clear cell RCC. To optimise these combination therapies while preserving patient quality of life, escalation and de-escalation strategies are being evaluated in prospective randomised trials, based on patient selection according to their prognosis risk. Finally, new therapeutic approaches, such as targeting hypoxia-inducible factor (HIF) and the development of innovative treatments using antibody-drug conjugates (ADCs), CAR-T cells, or radiopharmaceuticals, are all potential candidates to improve further patient survival.
Collapse
Affiliation(s)
- Mathieu Larroquette
- Department of Medical Oncology, University Hospital of Bordeaux, 33000 Bordeaux, France
- Faculty of Medicine, University of Bordeaux, 33000 Bordeaux, France
| | - Félix Lefort
- Department of Medical Oncology, University Hospital of Bordeaux, 33000 Bordeaux, France
| | - Luc Heraudet
- Department of Medical Oncology, University Hospital of Bordeaux, 33000 Bordeaux, France
- Faculty of Medicine, University of Bordeaux, 33000 Bordeaux, France
| | - Jean-Christophe Bernhard
- Faculty of Medicine, University of Bordeaux, 33000 Bordeaux, France
- Department of Urology, University Hospital of Bordeaux, 33000 Bordeaux, France
| | - Alain Ravaud
- Department of Medical Oncology, University Hospital of Bordeaux, 33000 Bordeaux, France
- Faculty of Medicine, University of Bordeaux, 33000 Bordeaux, France
| | - Charlotte Domblides
- Department of Medical Oncology, University Hospital of Bordeaux, 33000 Bordeaux, France
- Faculty of Medicine, University of Bordeaux, 33000 Bordeaux, France
| | - Marine Gross-Goupil
- Department of Medical Oncology, University Hospital of Bordeaux, 33000 Bordeaux, France
| |
Collapse
|
3
|
Di Bona C, Stühler V, Rausch S, Stenzl A, Bedke J. Pembrolizumab for the treatment of renal cell carcinoma. Expert Opin Biol Ther 2021; 21:1157-1164. [PMID: 34042015 DOI: 10.1080/14712598.2021.1935856] [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: 12/24/2022]
Abstract
INTRODUCTION The acquisition of resistance to VEGF-inhibiting therapies has prompted research toward immunotherapy for the treatment of metastatic renal cell carcinoma (mRCC). Among several, checkpoint inhibitors including PD-1 and PD-L1 inhibitors are the most promising approach. AREAS COVERED This review addresses the clinical efficacy of the anti-PD-1 monoclonal antibody pembrolizumab in first- and second-line treatment for mRCC regarding the most recent and significant published and ongoing studies. Attention is also given to its pharmacological characteristics as well as adverse events and its impact on patients' quality of life. EXPERT OPINION Immunotherapy has become the backbone for the treatment of advanced RCC. With the approval of several therapeutic options, research needs now to focus on defining the appropriate therapy for each patient. Axitinib plus pembrolizumab belongs to the combinations of tyrosine kinase inhibitors (TKI) plus immune checkpoint inhibitors for the first-line treatment of metastatic RCC. New combinations of pembrolizumab plus TKI for the evaluation in first- and second-line treatment of mRCC available. However, studies directly comparing the various treatment regimens using predictive biomarkers and long-term endpoints, including treatment-free survival, are lacking.
Collapse
Affiliation(s)
- Carlo Di Bona
- Department of Urology, University of Tübingen, Tuebingen, Germany
| | - Viktoria Stühler
- Department of Urology, University of Tübingen, Tuebingen, Germany
| | - Steffen Rausch
- Department of Urology, University of Tübingen, Tuebingen, Germany
| | - Arnulf Stenzl
- Department of Urology, University of Tübingen, Tuebingen, Germany
| | - Jens Bedke
- Department of Urology, University of Tübingen, Tuebingen, Germany
| |
Collapse
|
4
|
Hah YS, Koo KC. Immunology and Immunotherapeutic Approaches for Advanced Renal Cell Carcinoma: A Comprehensive Review. Int J Mol Sci 2021; 22:ijms22094452. [PMID: 33923219 PMCID: PMC8123195 DOI: 10.3390/ijms22094452] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 04/19/2021] [Accepted: 04/20/2021] [Indexed: 12/25/2022] Open
Abstract
Renal cell carcinoma (RCC) is a malignant tumor associated with various tumor microenvironments (TMEs). The immune system is activated by the development of cancer and drives T cell anti-tumor response. CD8 T cells are known to improve clinical outcomes and sensitivity to immunotherapy, and play a crucial role against tumors. In contrast, tumor-associated macrophages (TAMs) suppress immunity against malignancy and lead to tumor progression. TAMs are promoted from damaged TMEs and mount proinflammatory responses to pathogens. Initial immunotherapy consists of interferon-α and interleukin-2. However, response to such therapy is unclear in most patients, and it is associated with high levels of toxicity. Immune checkpoint inhibitors (ICIs), which up-regulate immune responses by blocking the programed cell death protein 1 (PD-1) receptor, the ligand of PD-1, or cytotoxic T-lymphocyte-associated protein 4 T cells, have led to a new era of immunotherapy. Furthermore, combination strategies with ICIs have proven effective through several randomized controlled trials. We expect the next generation of immunotherapy to lead to better outcomes based on ongoing trials and inspire new therapeutic strategies.
Collapse
Affiliation(s)
- Yoon-Soo Hah
- Department of Urology, Catholic University of Daegu School of Medicine, Daegu 42472, Korea;
| | - Kyo-Chul Koo
- Department of Urology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06229, Korea
- Correspondence: ; Tel.: +82-2-2019-3470
| |
Collapse
|
5
|
Loo V, Salgia M, Bergerot P, Philip EJ, Pal SK. First-Line Systemic Therapy for Metastatic Clear-Cell Renal Cell Carcinoma: Critical Appraisal of Emerging Options. Target Oncol 2020; 14:639-645. [PMID: 31595385 DOI: 10.1007/s11523-019-00676-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Until recently, a dichotomy existed in the front-line approach of metastatic clear-cell renal cell carcinoma (mRCC). Specifically, patients received either targeted therapy or immunotherapy. Targeted therapy entailed use of agents blocking signaling through the vascular endothelial growth factor (VEGF) receptor, such as cabozantinib, sunitinib, or pazopanib. Immunotherapy entailed dual therapy with nivolumab and ipilimumab, both checkpoint inhibitors for intermediate/poor International Metastatic RCC Database Consortium (IMDC)-risk disease patients. Within the past year, two datasets have emerged that led to recent approvals of combined therapy with VEGF and checkpoint inhibitors. These regimens (axitinib with either avelumab or pembolizumab) are among several that have been or will be evaluated for patients with newly diagnosed mRCC. We aim to facilitate treatment decisions through this comprehensive and contextualized overview of recent datasets in this therapeutic space.
Collapse
Affiliation(s)
- Vivian Loo
- Department of Protocol Content Administration, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Meghan Salgia
- Department of Medical Oncology and Experimental Therapeutics, City of Hope Comprehensive Cancer Center, 1500 East Duarte Road, Duarte, CA, 91010, USA
| | - Paulo Bergerot
- Department of Medical Oncology and Experimental Therapeutics, City of Hope Comprehensive Cancer Center, 1500 East Duarte Road, Duarte, CA, 91010, USA
| | | | - Sumanta K Pal
- Department of Medical Oncology and Experimental Therapeutics, City of Hope Comprehensive Cancer Center, 1500 East Duarte Road, Duarte, CA, 91010, USA.
| |
Collapse
|
6
|
Moreira M, Pobel C, Epaillard N, Simonaggio A, Oudard S, Vano YA. Resistance to cancer immunotherapy in metastatic renal cell carcinoma. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2020; 3:454-471. [PMID: 35582435 PMCID: PMC8992500 DOI: 10.20517/cdr.2020.16] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 05/06/2020] [Accepted: 05/29/2020] [Indexed: 12/17/2022]
Abstract
The prognosis of metastatic clear cell renal cell carcinoma (mccRCC) has changed dramatically over the years with the emergence of immune checkpoint inhibitors (ICI) used alone, or in combination with another ICI, or with vascular endothelial growth factor receptor tyrosine kinase inhibitor. Although major response rates have been observed with ICI, many patients do not respond, reflecting primary resistance, and durable responses remain exceptional, reflecting secondary resistance. Factors contributing to primary and acquired resistance are manifold, including patient-intrinsic factors, tumor cell-intrinsic factors and factors associated with the tumoral microenvironment (TME). While some mechanisms of resistance are common to several tumor types, others are specific to mccRCC. Predictive biomarkers and alternative strategies are needed to overcome this resistance. This review provides an overview of the major ICI resistance mechanisms, highlights the potential of the TME to induce resistance to ICI, and discusses the predictive biomarkers available to guide therapeutic choice.
Collapse
Affiliation(s)
- Marco Moreira
- Centre de Recherche des Cordeliers, INSERM, Université de Paris, Sorbonne Université, Paris F-75006, France.,Both authors contributed equally
| | - Cedric Pobel
- Department of Medical Oncology, European Hospital Georges Pompidou - APHP, Paris 75015, France.,University François Rabelais, Tours 37200, France.,Both authors contributed equally
| | - Nicolas Epaillard
- Department of Medical Oncology, European Hospital Georges Pompidou - APHP, Paris 75015, France
| | - Audrey Simonaggio
- Department of Medical Oncology, European Hospital Georges Pompidou - APHP, Paris 75015, France
| | - Stéphane Oudard
- Department of Medical Oncology, European Hospital Georges Pompidou - APHP, Paris 75015, France.,INSERM UMR-S1147, Paris 75006, France
| | - Yann-Alexandre Vano
- Centre de Recherche des Cordeliers, INSERM, Université de Paris, Sorbonne Université, Paris F-75006, France.,Department of Medical Oncology, European Hospital Georges Pompidou - APHP, Paris 75015, France
| |
Collapse
|
7
|
Liu X, Sun S, Liu D. IL-17D: A Less Studied Cytokine of IL-17 Family. Int Arch Allergy Immunol 2020; 181:618-623. [PMID: 32516792 DOI: 10.1159/000508255] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Accepted: 04/28/2020] [Indexed: 11/19/2022] Open
Abstract
The interleukin-17 (IL-17) family is a relatively new family of cytokines consisting of 6 related factors (IL-17A-IL-17F), while the receptor family consists of 5 members: IL-17RA-IL-17RE. IL-17A is the prototype member of this family, which is also the signature cytokine of T helper 17 (Th17) cells. Th17 cells are involved in the development of autoimmune disease, inflammation, and tumors. Although IL-17D is similar to IL-17A in its ability to induce inflammatory cytokine production, there are fewer studies on IL-17D. Recently, the role of IL-17D in tumors and infections has attracted our attention. Some knowledge of function of IL-17D has been gained by studies using nonmammalian species. In this review, we introduce the structural characteristics, expression patterns, and biological characteristics of IL-17D along with its potential function in the pathogenesis of disease.
Collapse
Affiliation(s)
- Xuying Liu
- Department of Gastroenterology and Medical Research Center, Liaoning Key Laboratory of Research and Application of Animal Models for Environmental and Metabolic Diseases, Sheng Jing Hospital of China Medical University, ShenYang, China
| | - Siyu Sun
- Department of Gastroenterology, ShengJing Hospital of China Medical University, ShenYang, China
| | - Dongyan Liu
- Department of Gastroenterology and Medical Research Center, Liaoning Key Laboratory of Research and Application of Animal Models for Environmental and Metabolic Diseases, Sheng Jing Hospital of China Medical University, ShenYang, China,
| |
Collapse
|
8
|
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
|
9
|
Intratumoral IFN-α gene delivery reduces tumor-infiltrating regulatory T cells through the downregulation of tumor CCL17 expression. Cancer Gene Ther 2018; 26:334-343. [PMID: 30420718 DOI: 10.1038/s41417-018-0059-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 10/12/2018] [Accepted: 10/20/2018] [Indexed: 01/13/2023]
Abstract
The effect of IFN-α on the immunosuppressive tumor microenvironment is not fully understood. We previously reported that intratumoral IFN-α gene transduction decreased the frequency of regulatory T cells (Tregs) in the tumor by inducing the secretion of IL-6 from dendritic cells. In this study, we examined whether IFN-α affects the trafficking of Tregs to the tumor. Since CT26 cells expressed CCL17 among Treg-attracting chemokines, we focused on its role in IFN-α-mediated Treg suppression. IFN-α directly suppressed CCL17 production from CT26 cells in vitro, and IFN-α transduction reduced CCL17 expression in tumors in vivo. Next, to investigate whether CCL17 downregulation is related to the suppression of Treg trafficking, CCL17-downregulated CT26 cells produced using short hairpin RNA (CT26-shCCL17) were inoculated into mice. The frequency of Tregs in CT26-shCCL17 tumors was reduced and tumor growth was suppressed. Finally, to examine the combinatorial effect of IFN-α expression with CCL17 downregulation, IFN-α was transduced into CT26-shCCL17 tumors. This resulted in an elevation of CT26-specific CD8+ T cells and the complete eradication of tumors. This study shows a novel mechanism of IFN-α-mediated Treg suppression, and combining IFN-α gene therapy with strong CCL17 downregulation could offer a promising strategy for the treatment of cancer.
Collapse
|
10
|
Ascierto PA, Brugarolas J, Buonaguro L, Butterfield LH, Carbone D, Daniele B, Ferris R, Fox BA, Galon J, Gridelli C, Kaufman HL, Klebanoff CA, Melero I, Nathan P, Paulos CM, Ruella M, Sullivan R, Zarour H, Puzanov I. Perspectives in immunotherapy: meeting report from the Immunotherapy Bridge (29-30 November, 2017, Naples, Italy). J Immunother Cancer 2018; 6:69. [PMID: 29996914 PMCID: PMC6042369 DOI: 10.1186/s40425-018-0377-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 06/19/2018] [Indexed: 12/26/2022] Open
Abstract
Immunotherapy represents the third important wave in the history of the systemic treatment of cancer after chemotherapy and targeted therapy and is now established as a potent and effective treatment option across several cancer types. The clinical success of anti-cytotoxic T-lymphocyte-associated antigen (CTLA)-4, first, and anti-programmed death (PD)-1/PD-ligand (L)1 agents in melanoma and other cancers a few years later, has encouraged increasing focus on the development of other immunotherapies (e.g. monoclonal antibodies with other immune targets, adoptive cell transfer, and vaccines), with over 3000 immuno-oncology trials ongoing, involving hundreds of research institutes across the globe. The potential use of these different immunotherapeutic options in various combinations with one another and with other treatment modalities is an area of particular promise. The third Immunotherapy Bridge meeting (29-30 November, 2017, Naples, Italy) focused on recent advances in immunotherapy across various cancer types and is summarised in this report.
Collapse
Affiliation(s)
- Paolo A. Ascierto
- Melanoma, Cancer Immunotherapy and Development Therapeutics Oncology Unit, Istituto Nazionale Tumori IRCCS Fondazione “G. Pascale, Napoli, Italy
| | - James Brugarolas
- Kidney Cancer Program, Department of Internal Medicine, Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas USA
| | - Luigi Buonaguro
- Molecular Biology and Viral Oncology Unit, Istituto Nazionale Tumori IRCCS Fondazione “G. Pascale, Napoli, Italy
| | - Lisa H. Butterfield
- UPCI Immunologic Monitoring and Cellular Products Laboratory, University of Pittsburgh, Pittsburgh, Pennsylvania USA
| | - David Carbone
- College of Medicine, James Thoracic Center, James Cancer Hospital and Solove Research Institute, The Ohio State University, Columbus, Ohio USA
| | - Bruno Daniele
- Department of Oncology, “G. Rummo” Hospital, Benevento, Italy
| | - Robert Ferris
- Division of Head and Neck Surgery, Department of Otolaryngology, University of Pittsburgh, Pittsburgh, Pennsylvania USA
| | - Bernard A. Fox
- Laboratory of Molecular and Tumor Immunology, Robert W. Franz Cancer Research Center in the Earle A. Chiles Research Institute at Providence Cancer Center, Portland, Oregon USA
| | - Jérôme Galon
- National Institute of Health and Medical Research (INSERM), Paris, France
| | - Cesare Gridelli
- Unit of Medical Oncology, Hospital “San Giuseppe Moscati”, Avellino, Italy
| | - Howard L. Kaufman
- Robert Wood Johnson Medical School Rutgers, The State University of New Jersey, New Brunswick, New Jersey USA
| | - Christopher A. Klebanoff
- Center for Cell Engineering and Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York USA
| | - Ignacio Melero
- Immunology and Immunotherapy Service, Clinica Universidad de Navarra, Pamplona, Navarra Spain
| | - Paul Nathan
- Mount Vernon Cancer Centre, Northwood, Middlesex UK
| | - Chrystal M. Paulos
- Department of Microbiology and Immunology Hollings Cancer Center, Medical University of South Carolina (MUSC), Charleston, South Carolina USA
| | - Marco Ruella
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, Pennsylvania USA
| | - Ryan Sullivan
- Medicine Harvard Medical School and Haematology/Oncology Department, Massachusetts General Hospital, Boston, Massachusetts USA
| | - Hassane Zarour
- Melanoma Program, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania USA
| | - Igor Puzanov
- Early Phase Clinical Trials Program, Experimental Therapeutics Program, Melanoma Section, Department of Medicine, Roswell Park Cancer Institute, Buffalo, New York USA
| |
Collapse
|
11
|
Li J, Sun J, Rong R, Li L, Shang W, Song D, Feng G, Luo F. HMGB1 promotes myeloid-derived suppressor cells and renal cell carcinoma immune escape. Oncotarget 2017; 8:63290-63298. [PMID: 28968989 PMCID: PMC5609921 DOI: 10.18632/oncotarget.18796] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 06/02/2017] [Indexed: 01/01/2023] Open
Abstract
Despite high immunogenicity and marked presence of immune cells in the RCC(renal cell carcinoma), immunotherapy fails to develop effective anti-tumor immune responses. This is due to the negative regulatory factors in the tumor microenvironment. As the main contributor of immunosuppression, myeloid-derived suppressor cells (MDSCs) inhibited anti-tumor immunity and promoted tumor progression. Meanwhile, it is confirmed that high mobility group box-1 protein (HMGB1) shows a high expression in many solid tumors and HMGB1 with high expression is involved in tumor immune escape. However, the mechanisms linking HMGB1 with tumor immune escape are unclear. The present study aimed to explore whether HMGB1 can promote RCC immune escape by inducing the generation of MDSCs. In this study, Renca mouse model was established and the influence of HMGB1 on MDSCs was investigated by using HMGB1 antibody to downregulate the expression of HMGB1 in tumor-bearing mice. The result showed that with the down-regulation of HMGB1, the tumor growth was inhibited significantly and the mice survival was prolonged greatly. Furthermore, the differentiation and proliferation of MDSCs were inhibited both in vitro and in vivo, and the inhibition rate showed a positive correlation with the degree of down-regulation of HMGB1. When MDSCs were eliminated with Gr-1 antibody in vivo, the ability of the HMGB1 to promote tumor growth was severely impaired. Thus, our findings indicated that HMGB1 might mediate tumor immune escape by promoting MDSCs cell proliferation, which provided a novel theoretical basis for preventing RCC using HMGB1 as the target.
Collapse
Affiliation(s)
- Jinfeng Li
- Kidney Transplantation Unit, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jiajia Sun
- Kidney Transplantation Unit, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ruiming Rong
- Department of Urology, Zhongshan Hospital and School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Long Li
- Department of Urology, Zhongshan Hospital and School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Wenjun Shang
- Kidney Transplantation Unit, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Dongkui Song
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Guiwen Feng
- Kidney Transplantation Unit, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Feifei Luo
- Department of Digestive Diseases, Huashan Hospital and Biotherapy Research Center, Fudan University, Shanghai, China
| |
Collapse
|
12
|
Affiliation(s)
- Toni K Choueiri
- From the Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Boston (T.K.C.); and the Department of Medicine, Genitourinary Oncology Service, Memorial Sloan Kettering Cancer Center, New York (R.J.M.)
| | - Robert J Motzer
- From the Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Boston (T.K.C.); and the Department of Medicine, Genitourinary Oncology Service, Memorial Sloan Kettering Cancer Center, New York (R.J.M.)
| |
Collapse
|
13
|
van den Brom R, van Es S, Leliveld A, Gietema J, Hospers G, de Jong I, de Vries E, Oosting S. Balancing treatment efficacy, toxicity and complication risk in elderly patients with metastatic renal cell carcinoma. Cancer Treat Rev 2016; 46:63-72. [DOI: 10.1016/j.ctrv.2016.04.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Revised: 04/06/2016] [Accepted: 04/08/2016] [Indexed: 11/15/2022]
|
14
|
Massari F, Santoni M, Ciccarese C, Santini D, Alfieri S, Martignoni G, Brunelli M, Piva F, Berardi R, Montironi R, Porta C, Cascinu S, Tortora G. PD-1 blockade therapy in renal cell carcinoma: current studies and future promises. Cancer Treat Rev 2015; 41:114-121. [PMID: 25586601 DOI: 10.1016/j.ctrv.2014.12.013] [Citation(s) in RCA: 139] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 12/30/2014] [Indexed: 10/24/2022]
Abstract
RCC is considered an immunogenic tumor with a prominent dysfunctional immune cell infiltrate, unable to control tumor growth. Evasion of immune surveillance, a process defined immune-editing, leads to malignant progression. The striking improvement of knowledge in immunology has led to the identification of immune checkpoints (such as CTLA-4 and PD-1), whose blockage enhances the antitumor immunity. The interaction between PD-1, an inducible inhibitory receptor expressed on lymphocytes and DCs, and PD-L1 ligand, expressed by tumor cells, results in a down-regulation of the T-cell response. Therefore, the PD-1/PD-L1 axis inhibition by targeted-antibodies, increasing the T-cell proliferation and cytotoxicity, represents a promising mechanism to stimulate the anti-tumor activity of the immune system, improving the outcomes of cancer patients. Several PD-1 and PD-L1 inhibitors have been evaluated in different tumor types, showing promising results. The interesting correlation between lymphocytes PD-1 expression and RCC advanced stage, grade and prognosis, as well as the selective PD-L1 expression by RCC tumor cells and its potential association with worse clinical outcomes, have led to the development of new anti PD-1/PD-L1 agents, alone or in combination with anti-angiogenic drugs or other immunotherapeutic approaches, for the treatment of RCC. In this review we discuss the role of PD-1/PD-L1 in RCC, focusing on the biological rationale, current clinical studies and promising therapeutic perspectives to target the PD-1 pathway.
Collapse
Affiliation(s)
- F Massari
- Medical Oncology, Azienda Ospedaliera Universitaria Integrata, University of Verona, Piazzale L.A. Scuro 10, 37124 Verona, Italy
| | - M Santoni
- Clinica di Oncologia Medica, AOU "Ospedali Riuniti", Polytechnic University of the Marche Region, via Conca 71, 60126 Ancona, Italy.
| | - C Ciccarese
- Medical Oncology, Azienda Ospedaliera Universitaria Integrata, University of Verona, Piazzale L.A. Scuro 10, 37124 Verona, Italy
| | - D Santini
- Department of Medical Oncology, University Campus Bio-Medico Roma, Oncologia Medica, Rome, Italy
| | - S Alfieri
- SSD Oncologia medica Tumori Testa e Collo, Fondazione IRCCS - Istituto Nazionale dei Tumori, Via Venezian, 1, 20133 Milano, Italy
| | - G Martignoni
- Department of Pathology and Diagnostic, Azienda Ospedaliera Universitaria Integrata, University of Verona, Piazzale L.A. Scuro 10, 37124 Verona, Italy
| | - M Brunelli
- Department of Pathology and Diagnostic, Azienda Ospedaliera Universitaria Integrata, University of Verona, Piazzale L.A. Scuro 10, 37124 Verona, Italy
| | - F Piva
- Department of Specialistic Clinical and Odontostomatological Sciences, Polytechnic University of Marche, 60131 Ancona, Italy
| | - R Berardi
- Clinica di Oncologia Medica, AOU "Ospedali Riuniti", Polytechnic University of the Marche Region, via Conca 71, 60126 Ancona, Italy
| | - R Montironi
- Section of Pathological Anatomy, Polytechnic University of the Marche Region, School of Medicine, United Hospitals, Ancona, Italy
| | - C Porta
- Medical Oncology, IRCCS San Matteo University Hospital Foundation, piazzale C. Golgi 19, 27100 Pavia, Italy
| | - S Cascinu
- Clinica di Oncologia Medica, AOU "Ospedali Riuniti", Polytechnic University of the Marche Region, via Conca 71, 60126 Ancona, Italy
| | - G Tortora
- Medical Oncology, Azienda Ospedaliera Universitaria Integrata, University of Verona, Piazzale L.A. Scuro 10, 37124 Verona, Italy
| |
Collapse
|
15
|
Bielecka ZF, Czarnecka AM, Szczylik C. Genomic Analysis as the First Step toward Personalized Treatment in Renal Cell Carcinoma. Front Oncol 2014; 4:194. [PMID: 25120953 PMCID: PMC4110478 DOI: 10.3389/fonc.2014.00194] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Accepted: 07/09/2014] [Indexed: 12/13/2022] Open
Abstract
Drug resistance mechanisms in renal cell carcinoma (RCC) still remain elusive. Although most patients initially respond to targeted therapy, acquired resistance can still develop eventually. Most of the patients suffer from intrinsic (genetic) resistance as well, suggesting that there is substantial need to broaden our knowledge in the field of RCC genetics. As molecular abnormalities occur for various reasons, ranging from single nucleotide polymorphisms to large chromosomal defects, conducting whole-genome association studies using high-throughput techniques seems inevitable. In principle, data obtained via genome-wide research should be continued and performed on a large scale for the purposes of drug development and identification of biological pathways underlying cancerogenesis. Genetic alterations are mostly unique for each histological RCC subtype. According to recently published data, RCC is a highly heterogeneous tumor. In this paper, the authors discuss the following: (1) current state-of-the-art knowledge on the potential biomarkers of RCC subtypes; (2) significant obstacles encountered in the translational research on RCC; and (3) recent molecular findings that may have a crucial impact on future therapeutic approaches.
Collapse
Affiliation(s)
- Zofia Felicja Bielecka
- Department of Oncology with the Laboratory of Molecular Oncology, Military Institute of Medicine , Warsaw , Poland ; Postgraduate School of Molecular Medicine, Medical University of Warsaw , Warsaw , Poland
| | - Anna Małgorzata Czarnecka
- Department of Oncology with the Laboratory of Molecular Oncology, Military Institute of Medicine , Warsaw , Poland
| | - Cezary Szczylik
- Department of Oncology with the Laboratory of Molecular Oncology, Military Institute of Medicine , Warsaw , Poland
| |
Collapse
|
16
|
O'Sullivan T, Saddawi-Konefka R, Gross E, Tran M, Mayfield SP, Ikeda H, Bui JD. Interleukin-17D mediates tumor rejection through recruitment of natural killer cells. Cell Rep 2014; 7:989-98. [PMID: 24794441 DOI: 10.1016/j.celrep.2014.03.073] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2012] [Revised: 01/08/2014] [Accepted: 03/31/2014] [Indexed: 01/02/2023] Open
Abstract
The process of cancer immunoediting generates a repertoire of cancer cells that can persist in immune-competent hosts. In its most complex form, this process begins with the elimination of highly immunogenic unedited tumor cells followed by the escape of less immunogenic edited cells. Although edited tumors can release immunosuppressive factors, it is unknown whether unedited tumors produce cytokines that enhance antitumor function. Utilizing gene microarray analysis, we found the cytokine interleukin 17D (IL-17D) was highly expressed in certain unedited tumors but not in edited mouse tumor cell lines. Moreover, forced expression of IL-17D in edited tumor cells induced rejection by stimulating MCP-1 production from tumor endothelial cells, leading to the recruitment of natural killer (NK) cells. NK cells promoted M1 macrophage development and adaptive immune responses. IL-17D expression was also decreased in certain high-grade and metastatic human tumors, suggesting that it can be targeted for tumor immune therapy.
Collapse
Affiliation(s)
- Timothy O'Sullivan
- Department of Pathology, University of California, San Diego, 9500 Gilman Drive MC 0612, La Jolla, CA 92093, USA
| | - Robert Saddawi-Konefka
- Department of Pathology, University of California, San Diego, 9500 Gilman Drive MC 0612, La Jolla, CA 92093, USA
| | - Emilie Gross
- Department of Pathology, University of California, San Diego, 9500 Gilman Drive MC 0612, La Jolla, CA 92093, USA
| | - Miller Tran
- Department of Biology, University of California, San Diego, 9500 Gilman Drive MC 0612, La Jolla, CA 92093, USA
| | - Stephen P Mayfield
- Department of Biology, University of California, San Diego, 9500 Gilman Drive MC 0612, La Jolla, CA 92093, USA
| | - Hiroaki Ikeda
- Department of Immuno-Gene Therapy, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie 514-8507, Japan
| | - Jack D Bui
- Department of Pathology, University of California, San Diego, 9500 Gilman Drive MC 0612, La Jolla, CA 92093, USA.
| |
Collapse
|
17
|
Santoni M, De Tursi M, Felici A, Lo Re G, Ricotta R, Ruggeri EM, Sabbatini R, Santini D, Vaccaro V, Milella M. Management of metastatic renal cell carcinoma patients with poor-risk features: current status and future perspectives. Expert Rev Anticancer Ther 2014; 13:697-709. [PMID: 23773104 DOI: 10.1586/era.13.52] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
With seven agents approved for renal cell carcinoma within the past few years, there has undoubtedly been progress in treating this disease. However, patients with poor-risk features remain a challenging and difficult-to-treat population, with the mTOR inhibitor, temsirolimus, the only agent approved in the first-line setting. Phase III trial data are still lacking VEGF-pathway inhibitors in patients with poor prognostic features. Poor-risk patients need to be considered as a heterogeneous population. Further understanding of biomarkers can lead to a better selection of patients who may benefit the most from treatment and improvements in prognosis. The presence of poor Karnofsky scores and liver or CNS disease may affect the outcome of these patients much more than other identified factors. This consideration may provide the rationale to further stratify poor-risk patients further subgroups destined to receive either cure or palliation.
Collapse
Affiliation(s)
- Matteo Santoni
- Clinica di Oncologia Medica, AOU Ospedali Riuniti, Università Politecnica delle Marche, Ancona, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Aida K, Miyakawa R, Suzuki K, Narumi K, Udagawa T, Yamamoto Y, Chikaraishi T, Yoshida T, Aoki K. Suppression of Tregs by anti-glucocorticoid induced TNF receptor antibody enhances the antitumor immunity of interferon-α gene therapy for pancreatic cancer. Cancer Sci 2014; 105:159-67. [PMID: 24289533 PMCID: PMC4317823 DOI: 10.1111/cas.12332] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2013] [Revised: 11/25/2013] [Accepted: 11/27/2013] [Indexed: 12/29/2022] Open
Abstract
We have reported that interferon (IFN)-α can attack cancer cells by multiple antitumor mechanisms including the induction of direct cancer cell death and the enhancement of an immune response in several pancreatic cancer models. However, an immunotolerant microenvironment in the tumors is often responsible for the failure of the cancer immunotherapy. Here we examined whether the suppression of regulatory T cells (Tregs) within tumors can enhance an antitumor immunity induced by an intratumoral IFN-α gene transfer. First we showed that an intraperitoneal administration of an agonistic anti-glucocorticoid induced TNF receptor (GITR) monoclonal antibody (mAb), which is reported to suppress the function of Tregs, significantly inhibited subcutaneous tumor growth in a murine pancreatic cancer model. The anti-GITR mAb was then combined with the intratumoral injection of the IFN-α-adenovirus vector. The treatment with the antibody synergistically augmented the antitumor effect of IFN-α gene therapy not only in the vector-injected tumors but also in the vector-uninjected tumors. Immunostaining showed that the anti-GITR mAb decreased Foxp3+ cells infiltrating in the tumors, while the intratumoral IFN-α gene transfer increased CD4+ and CD8+ T cells in the tumors. Therefore, the combination therapy strongly inclined the immune balance of the tumor microenvironment in an antitumor direction, leading to a marked systemic antitumor effect. The CCR5 expression on Tregs was downregulated in the antibody-treated mice, which may explain the decrease of tumor-infiltrating Tregs. The combination of Treg-suppression by GITR mAb and the tumor immunity induction by IFN-α gene therapy could be a promising therapeutic strategy for pancreatic cancer.
Collapse
Affiliation(s)
- Kouichirou Aida
- Division of Gene and Immune Medicine, National Cancer Center Research Institute, Tokyo, Japan; Department of Urology, St. Marianna University, Kanagawa, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Thapa RJ, Chen P, Cheung M, Nogusa S, Pei J, Peri S, Testa JR, Balachandran S. NF-κB inhibition by bortezomib permits IFN-γ-activated RIP1 kinase-dependent necrosis in renal cell carcinoma. Mol Cancer Ther 2013; 12:1568-78. [PMID: 23657944 DOI: 10.1158/1535-7163.mct-12-1010] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Advanced renal cell carcinoma (RCC) is an invariably fatal cancer. Currently, small-molecule inhibitors that target cell growth, angiogenesis, or nutrient-sensing pathways represent the primary pharmacologic interventions for this disease, but these inhibitors only delay tumor progression and are not curative. The cytokine IFN-γ showed the potential to provide lasting remission in several phase I/II trials for advanced RCCs, but subsequent trials, including a multicenter phase III study using IFN-γ as a monotherapy for RCCs, were less promising. Notably, these trials were designed to exploit the indirect immunomodulatory effects of IFN-γ, whereas its direct antitumor properties--including its ability to trigger programmed cell death in tumors-remain mostly untapped. Here, we show that the proteasome inhibitor bortezomib (PS-341, Velcade) sensitizes otherwise resistant RCC cells to direct necrotic death by IFN-γ. Mechanistically, we show that bortezomib functions, at least in part, by inhibiting prosurvival NF-κB signaling. In the absence of this signal, IFN-γ triggers programmed necrosis (or "necroptosis") dependent on the kinase RIP1. When taken together with the observation that NF-κB signaling is elevated in RCCs, these results provide rationale for the combined use of IFN-γ and bortezomib in the treatment of metastatic RCCs.
Collapse
Affiliation(s)
- Roshan J Thapa
- Immune Cell Development and Host Defense Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Chen P, Nogusa S, Thapa RJ, Shaller C, Simmons H, Peri S, Adams GP, Balachandran S. Anti-CD70 immunocytokines for exploitation of interferon-γ-induced RIP1-dependent necrosis in renal cell carcinoma. PLoS One 2013; 8:e61446. [PMID: 23613854 PMCID: PMC3629199 DOI: 10.1371/journal.pone.0061446] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Accepted: 03/09/2013] [Indexed: 01/19/2023] Open
Abstract
Metastatic renal cell carcinoma (RCC) is an incurable disease in clear need of new therapeutic interventions. In early-phase clinical trials, the cytokine IFN-γ showed promise as a biotherapeutic for advanced RCC, but subsequent trials were less promising. These trials, however, focused on the indirect immunomodulatory properties of IFN-γ, and its direct anti-tumor effects, including its ability to kill tumor cells, remains mostly unexploited. We have previously shown that IFN-γ induces RIP1 kinase-dependent necrosis in cells lacking NF-κB survival signaling. RCC cells display basally-elevated NF-κB activity, and inhibiting NF-κB in these cells, for example by using the small-molecule proteasome blocker bortezomib, sensitizes them to RIP1-dependent necrotic death following exposure to IFN-γ. While these observations suggest that IFN-γ-mediated direct tumoricidal activity will have therapeutic benefit in RCC, they cannot be effectively exploited unless IFN-γ is targeted to tumor cells in vivo. Here, we describe the generation and characterization of two novel ‘immunocytokine’ chimeric proteins, in which either human or murine IFN-γ is fused to an antibody targeting the putative metastatic RCC biomarker CD70. These immunocytokines display high levels of species-specific IFN-γ activity and selective binding to CD70 on human RCC cells. Importantly, the IFN-γ immunocytokines function as well as native IFN-γ in inducing RIP1-dependent necrosis in RCC cells, when deployed in the presence of bortezomib. These results provide a foundation for the in vivo exploitation of IFN-γ-driven tumoricidal activity in RCC.
Collapse
Affiliation(s)
- Peirong Chen
- Immune Cell Development and Host Defense Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania, United States of America
| | - Shoko Nogusa
- Immune Cell Development and Host Defense Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania, United States of America
| | - Roshan J. Thapa
- Immune Cell Development and Host Defense Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania, United States of America
| | - Calvin Shaller
- Developmental Therapeutics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania, United States of America
| | - Heidi Simmons
- Developmental Therapeutics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania, United States of America
| | - Suraj Peri
- Department of Biostatistics and Bioinformatics, Fox Chase Cancer Center, Philadelphia, Pennsylvania, United States of America
| | - Gregory P. Adams
- Developmental Therapeutics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania, United States of America
| | - Siddharth Balachandran
- Immune Cell Development and Host Defense Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
21
|
|
22
|
Inman BA, Harrison MR, George DJ. Novel immunotherapeutic strategies in development for renal cell carcinoma. Eur Urol 2012; 63:881-9. [PMID: 23084331 DOI: 10.1016/j.eururo.2012.10.006] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2012] [Accepted: 10/05/2012] [Indexed: 01/08/2023]
Abstract
CONTEXT The purpose of this report is to review immunotherapies under investigation for patients with renal cell carcinoma (RCC), the most common form of kidney cancer, for which the incidence and mortality rate continue to increase. OBJECTIVE To summarize and evaluate current data on immunotherapies for RCC and discuss issues to be resolved before integration into the RCC treatment paradigm. EVIDENCE ACQUISITION A search of Medline, clinicaltrials.gov, and congress abstracts/treatment guidelines was performed in May 2012 using the following terms (and variations): metastatic renal cell carcinoma, practice guidelines, response/resistance to current treatments, immunotherapy, novel immunotherapeutic strategies, T-cell modulation, immune priming, innate immunity, and combination therapy. EVIDENCE SYNTHESIS Prior to the advent of novel agents targeting the vascular endothelial growth factor and mechanistic target of rapamycin pathways, interleukin-2 (IL-2) and interferon-α were the mainstays of RCC treatment. IL-2 remains one of the only treatments capable of curing advanced RCC, albeit in few patients. Despite recent advances, unmet need still exists for patients in the adjuvant setting, those with poor prognostic factors, and those who have progressed on prior targeted therapies. Improved understanding of host-tumor immune interactions has led to development of novel immunotherapeutic agents, including antibodies against immune checkpoint proteins (eg, programmed death-1 and cytotoxic T-lymphocyte antigen-4), and various vaccines. Because many of these compounds are in development, clinical experience with them is limited, although some have demonstrated activity in preliminary studies. CONCLUSIONS It is not yet clear where these new immunotherapies will fit into RCC treatment paradigms, but they may provide new options for patients whose current choices are limited. Furthermore, predictive biomarkers are needed to identify patients who will derive the greatest benefit from immunotherapy.
Collapse
Affiliation(s)
- Brant A Inman
- Division of Urology, Duke University Medical Center, Durham, NC 27710, USA.
| | | | | |
Collapse
|
23
|
Abstract
Immunotherapy has been used for the treatment of renal diseases for a long time, and there has been significant progress in such treatment. This review focuses on the use of immunotherapy for the treatment of glomerular diseases. The use of immunosuppression in the treatment of minimal change disease, membranous nephropathy, primary focal segmental glomerulosclerosis, lupus nephritis, immunoglobulin-A nephropathy, antineutrophil cytoplasmic antibody-associated disease, and anti-glomerular basement membrane disease is discussed.
Collapse
Affiliation(s)
- Ajay Kher
- The Transplant Institute, Beth Israel Deaconess Medical Center, 110 Francis Street, 7th Floor, Boston, MA 02215, USA.
| | | |
Collapse
|