6001
|
Johnson DB, Estrada MV, Salgado R, Sanchez V, Doxie DB, Opalenik SR, Vilgelm AE, Feld E, Johnson AS, Greenplate AR, Sanders ME, Lovly CM, Frederick DT, Kelley MC, Richmond A, Irish JM, Shyr Y, Sullivan RJ, Puzanov I, Sosman JA, Balko JM. Melanoma-specific MHC-II expression represents a tumour-autonomous phenotype and predicts response to anti-PD-1/PD-L1 therapy. Nat Commun 2016; 7:10582. [PMID: 26822383 PMCID: PMC4740184 DOI: 10.1038/ncomms10582] [Citation(s) in RCA: 428] [Impact Index Per Article: 47.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 12/31/2015] [Indexed: 12/18/2022] Open
Abstract
Anti-PD-1 therapy yields objective clinical responses in 30-40% of advanced melanoma patients. Since most patients do not respond, predictive biomarkers to guide treatment selection are needed. We hypothesize that MHC-I/II expression is required for tumour antigen presentation and may predict anti-PD-1 therapy response. In this study, across 60 melanoma cell lines, we find bimodal expression patterns of MHC-II, while MHC-I expression was ubiquitous. A unique subset of melanomas are capable of expressing MHC-II under basal or IFNγ-stimulated conditions. Using pathway analysis, we show that MHC-II(+) cell lines demonstrate signatures of 'PD-1 signalling', 'allograft rejection' and 'T-cell receptor signalling', among others. In two independent cohorts of anti-PD-1-treated melanoma patients, MHC-II positivity on tumour cells is associated with therapeutic response, progression-free and overall survival, as well as CD4(+) and CD8(+) tumour infiltrate. MHC-II(+) tumours can be identified by melanoma-specific immunohistochemistry using commercially available antibodies for HLA-DR to improve anti-PD-1 patient selection.
Collapse
Affiliation(s)
- Douglas B. Johnson
- Department of Medicine, Vanderbilt University, Nashville, 37232 Tennessee, USA,
| | - Monica V. Estrada
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, 37232 Tennessee, USA
| | - Roberto Salgado
- Department of Pathology, Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Boulevard de Waterloo 121, Brussels 1000, Belgium
| | - Violeta Sanchez
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, 37232 Tennessee, USA
| | - Deon B. Doxie
- Department of Cancer Biology, Vanderbilt University, Nashville, 37232 Tennessee, USA
| | - Susan R. Opalenik
- Department of Medicine, Vanderbilt University, Nashville, 37232 Tennessee, USA
| | - Anna E. Vilgelm
- Department of Cancer Biology, Vanderbilt University, Nashville, 37232 Tennessee, USA,Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, 37232 Tennessee, USA
| | - Emily Feld
- Department of Medicine, Vanderbilt University, Nashville, 37232 Tennessee, USA
| | - Adam S. Johnson
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, 37232 Tennessee, USA
| | - Allison R. Greenplate
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, 37232 Tennessee, USA,Department of Cancer Biology, Vanderbilt University, Nashville, 37232 Tennessee, USA
| | - Melinda E. Sanders
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, 37232 Tennessee, USA
| | - Christine M. Lovly
- Department of Medicine, Vanderbilt University, Nashville, 37232 Tennessee, USA,Department of Cancer Biology, Vanderbilt University, Nashville, 37232 Tennessee, USA
| | - Dennie T. Frederick
- Department of Medicine, Massachusetts General Hospital, Boston, 02114 Massachusetts, USA
| | - Mark C. Kelley
- Department of Surgical Oncology, Vanderbilt University, Nashville, 37232 Tennessee, USA
| | - Ann Richmond
- Department of Cancer Biology, Vanderbilt University, Nashville, 37232 Tennessee, USA,Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, 37232 Tennessee, USA
| | - Jonathan M. Irish
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, 37232 Tennessee, USA,Department of Cancer Biology, Vanderbilt University, Nashville, 37232 Tennessee, USA
| | - Yu Shyr
- Department of Biostatistics, Vanderbilt University, Nashville, 37232 Tennessee, USA
| | - Ryan J. Sullivan
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, 37232 Tennessee, USA
| | - Igor Puzanov
- Department of Medicine, Vanderbilt University, Nashville, 37232 Tennessee, USA
| | - Jeffrey A. Sosman
- Department of Medicine, Vanderbilt University, Nashville, 37232 Tennessee, USA
| | - Justin M. Balko
- Department of Medicine, Vanderbilt University, Nashville, 37232 Tennessee, USA,Department of Cancer Biology, Vanderbilt University, Nashville, 37232 Tennessee, USA,
| |
Collapse
|
6002
|
Eke I, Makinde AY, Aryankalayil MJ, Ahmed MM, Coleman CN. Comprehensive molecular tumor profiling in radiation oncology: How it could be used for precision medicine. Cancer Lett 2016; 382:118-126. [PMID: 26828133 DOI: 10.1016/j.canlet.2016.01.041] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 01/21/2016] [Accepted: 01/26/2016] [Indexed: 12/16/2022]
Abstract
New technologies enabling the analysis of various molecules, including DNA, RNA, proteins and small metabolites, can aid in understanding the complex molecular processes in cancer cells. In particular, for the use of novel targeted therapeutics, elucidation of the mechanisms leading to cell death or survival is crucial to eliminate tumor resistance and optimize therapeutic efficacy. While some techniques, such as genomic analysis for identifying specific gene mutations or epigenetic testing of promoter methylation, are already in clinical use, other "omics-based" assays are still evolving. Here, we provide an overview of the current status of molecular profiling methods, including promising research strategies, as well as possible challenges, and their emerging role in radiation oncology.
Collapse
Affiliation(s)
- Iris Eke
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Adeola Y Makinde
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Molykutty J Aryankalayil
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Mansoor M Ahmed
- Radiation Research Program, National Cancer Institute, National Institutes of Health, Rockville, MD 20850, USA
| | - C Norman Coleman
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; Radiation Research Program, National Cancer Institute, National Institutes of Health, Rockville, MD 20850, USA
| |
Collapse
|
6003
|
Perez-Chanona E, Trinchieri G. The role of microbiota in cancer therapy. Curr Opin Immunol 2016; 39:75-81. [PMID: 26820225 DOI: 10.1016/j.coi.2016.01.003] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 01/07/2016] [Indexed: 02/06/2023]
Abstract
The relationship between the host and the commensal microbiota regulates physiological functions including inflammation and immunity and it has been scrutinized in the context of cancer. While viruses and bacterial species have been implicated in oncogenesis, commensal microbes also have a beneficial role in the fight against cancer. Therapy efficacy, including adoptive T cell transfer, alkylating agents and immune checkpoint blockers, relies on immunity that receives its education from the gut microbiota. In cancer therapy with immunostimulating oligonucleotides and platinum salts, the microbiota also modulates the response by priming for the release of pro-inflammatory factors and reactive oxygen species, respectively. This new information offers promising clinical possibilities of modulating cancer therapy and its toxic side effects by targeting the microbiota.
Collapse
Affiliation(s)
- Ernesto Perez-Chanona
- Cancer and Inflammation Program, National Cancer Institute, Bethesda, MD 20892, USA; Leidos Biomedical Research, Inc., Frederick, MD 21701, USA
| | - Giorgio Trinchieri
- Cancer and Inflammation Program, National Cancer Institute, Bethesda, MD 20892, USA.
| |
Collapse
|
6004
|
Suzuki S, Ishida T, Yoshikawa K, Ueda R. Current status of immunotherapy. Jpn J Clin Oncol 2016; 46:191-203. [PMID: 26819277 DOI: 10.1093/jjco/hyv201] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 12/12/2015] [Indexed: 02/07/2023] Open
Abstract
The successful use of immune checkpoint inhibitors has been big breakthrough in the development of cancer immunotherapy. Anti-CTLA-4 monoclonal antibody, ipilimumab, is the first-approved immune checkpoint inhibitor and has shown durable objective responses for advanced melanoma beyond the effect of dacarbazine. Anti-PD-1 monoclonal antibodies, nivolumab and pembrolizumab, are other immune checkpoint inhibitors that have demonstrated more effective results than conventional drugs in clinical trials for a variety of advanced solid tumors including melanoma, non-small cell lung carcinoma and renal carcinoma. These studies have indicated that the enhancement of anti-cancer immunity by controlling the immune suppressive environment in cancer tissues is an important issue for the development of cancer immune-therapy. Accordingly, in recent years, the enthusiasm for research of cancer immunology has shifted to studies regarding the formation of the immune suppressive environment, immune suppression mechanisms in cancer tissues and the molecules and cells involved in these pathways. Novel findings from these studies might lead to the development of cancer immunotherapy based on control of the immune suppressive environment.
Collapse
Affiliation(s)
- Susumu Suzuki
- Department of Tumor Immunology, Aichi Medical University School of Medicine, Nagakute
| | - Takashi Ishida
- Department of Hematology and Oncology, Nagoya City University Graduate School of Medical Sciences, Mizuho-ku, Nagoya
| | - Kazuhiro Yoshikawa
- Center for Advanced Medical Research, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| | - Ryuzo Ueda
- Department of Tumor Immunology, Aichi Medical University School of Medicine, Nagakute
| |
Collapse
|
6005
|
Abstract
As calculated by the meta-analysis of Korn et al., the prognosis of metastatic melanoma in the pretarget and immunological therapy era was poor, with a median survival of 6.2 and a 1-year life expectancy of 25.5%. Nowadays, significant advances in melanoma treatment have been gained, and immunotherapy is one of the promising approaches to get to durable responses and survival improvement. The aim of the present review is to highlight the recent innovations in melanoma immunotherapy and to propose a critical perspective of the future directions of this enthralling oncology subspecialty.
Collapse
Affiliation(s)
- Sara Valpione
- Christie Hospital NHS Foundation Trust, 550 Wilmslow Rd, Manchester, M20 4BX, UK
| | - Luca G Campana
- Department of Surgery, Oncology & Gastroenterology, University of Padova, via Gattamelata 64, 35128 Padova, Italy
| |
Collapse
|
6006
|
Jing W, Li M, Zhang Y, Teng F, Han A, Kong L, Zhu H. PD-1/PD-L1 blockades in non-small-cell lung cancer therapy. Onco Targets Ther 2016; 9:489-502. [PMID: 26889087 PMCID: PMC4741366 DOI: 10.2147/ott.s94993] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Lung cancer is the leading cause of cancer death in males and the second leading cause of death in females worldwide. Non-small-cell lung cancer (NSCLC) is the main pathological type of lung cancer, and most newly diagnosed NSCLC patients cannot undergo surgery because the disease is already locally advanced or metastatic. Despite chemoradiotherapy and targeted therapy improving clinical outcomes, overall survival remains poor. Immune checkpoint blockade, especially blockade of programmed death-1 (PD-1) receptor and its ligand PD-L1, achieved robust responses and improved survival for patients with locally advanced/metastatic NSCLC in preclinical and clinical studies. However, with regard to PD-1/PD-L1 checkpoint blockade as monotherapy or in combination with other antitumor therapies, such as chemotherapy, radiotherapy (including conventional irradiation and stereotactic body radiotherapy), and target therapy, there are still many unknowns in treating patients with NSCLC. Despite this limited understanding, checkpoint blockade as a novel therapeutic approach may change the treatment paradigm of NSCLC in the future. Here we review the main results from completed and ongoing studies to investigate the feasibility of PD-1/PD-L1 inhibitors, as monotherapy or combinatorial agents in patients with locally advanced and metastatic NSCLC, and explore optimal strategy in such patients.
Collapse
Affiliation(s)
- Wang Jing
- Weifang Medical University, Weifang, Shandong Province, People’s Republic of China
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Jinan, Shandong Province, People’s Republic of China
| | - Miaomiao Li
- Shandong Medical College, Jinan, Shandong Province, People’s Republic of China
| | - Yan Zhang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Jinan, Shandong Province, People’s Republic of China
| | - Feifei Teng
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Jinan, Shandong Province, People’s Republic of China
| | - Anqin Han
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Jinan, Shandong Province, People’s Republic of China
| | - Li Kong
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Jinan, Shandong Province, People’s Republic of China
| | - Hui Zhu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Jinan, Shandong Province, People’s Republic of China
| |
Collapse
|
6007
|
Wei XX, Fong L, Small EJ. Prospects for the use of ipilimumab in treating advanced prostate cancer. Expert Opin Biol Ther 2016; 16:421-32. [PMID: 26698365 DOI: 10.1517/14712598.2016.1136284] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Ipilimumab is a fully human monoclonal antibody that blocks Cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) to potentiate antitumor T cell response. Ipilimumab is approved for the treatment of advanced melanoma based on improved overall survival. Clinical trials of ipilimumab in patients with metastatic castrate-resistant prostate cancer (mCRPC) have demonstrated some clinical activity, but have largely been disappointing to date. AREAS COVERED Results of key clinical studies of ipilimumab in the treatment of prostate cancer, including clinical efficacy and toxicities, are summarized. EXPERT OPINION There is likely a clinical benefit to ipilimumab in a subset of mCRPC patients. The development of biomarkers for enrichment treatment strategies that select for patients most likely to benefit from ipilimumab is a top priority. Similarly, an understanding of the factors predictive of toxicity will be important in the development of future treatment approaches.
Collapse
Affiliation(s)
- Xiao X Wei
- a Division of Hematology and Oncology, UCSF Helen Diller Family Comprehensive Cancer Center , University of California, San Francisco , San Francisco , CA , USA
| | - Lawrence Fong
- a Division of Hematology and Oncology, UCSF Helen Diller Family Comprehensive Cancer Center , University of California, San Francisco , San Francisco , CA , USA
| | - Eric J Small
- a Division of Hematology and Oncology, UCSF Helen Diller Family Comprehensive Cancer Center , University of California, San Francisco , San Francisco , CA , USA
| |
Collapse
|
6008
|
Long GV, Weber JS, Infante JR, Kim KB, Daud A, Gonzalez R, Sosman JA, Hamid O, Schuchter L, Cebon J, Kefford RF, Lawrence D, Kudchadkar R, Burris HA, Falchook GS, Algazi A, Lewis K, Puzanov I, Ibrahim N, Sun P, Cunningham E, Kline AS, Del Buono H, McDowell DO, Patel K, Flaherty KT. Overall Survival and Durable Responses in Patients With BRAF V600-Mutant Metastatic Melanoma Receiving Dabrafenib Combined With Trametinib. J Clin Oncol 2016; 34:871-8. [PMID: 26811525 DOI: 10.1200/jco.2015.62.9345] [Citation(s) in RCA: 231] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE To report the overall survival (OS) and clinical characteristics of BRAF inhibitor-naive long-term responders and survivors treated with dabrafenib plus trametinib in a phase I and II study of patients with BRAF V600 mutation-positive metastatic melanoma. METHODS BRAF inhibitor-naive patients treated with dabrafenib 150 mg twice daily plus trametinib 2 mg daily (the 150/2 group) from the non-randomly assigned (part B) and randomly assigned (part C) cohorts of the study were analyzed for progression-free and OS separately. Baseline characteristics and factors on treatment were analyzed for associations with durable responses and OS. RESULTS For BRAF inhibitor-naive patients in the 150/2 groups (n = 78), the progression-free survival at 1, 2, and 3 years was 44%, 22%, and 18%, respectively, for part B (n = 24) and 41%, 25%, and 21%, respectively, for part C (n = 54). Median OS was 27.4 months in part B and 25 months in part C. OS at 1, 2, and 3 years was 72%, 60%, and 47%, respectively, for part B and 80%, 51%, and 38%, respectively, for part C. Prolonged survival was associated with metastases in fewer than three organ sites and lower baseline lactate dehydrogenase. OS at 3 years was 62% in patients with normal baseline lactate dehydrogenase and 63% in patients with a complete response. CONCLUSION Dabrafenib plus trametinib results in a median OS of more than 2 years in BRAF inhibitor-naive patients with BRAF V600 mutation-positive metastatic melanoma, and approximately 20% were progression free at 3 years. Durable responses occurred in patients with good prognostic features at baseline, which may be predictive.
Collapse
Affiliation(s)
- Georgina V Long
- Georgina V. Long, Melanoma Institute Australia; The University of Sydney; Richard F. Kefford, Melanoma Institute Australia; The University of Sydney; Macquarie University, Sydney; Westmead Hospital, Westmead; Jonathan Cebon, Austin Health, Melbourne, Victoria, Australia; Jeffrey S. Weber and Ragini Kudchadkar, Moffitt Cancer Center, Tampa, FL; Jeffrey R. Infante and Howard A. Burris III, Sarah Cannon Research Institute/Tennessee Oncology; Kevin B. Kim, California Pacific Medical Center; Adil Daud, Alain Algazi, University of California, San Francisco, San Francisco; Omid Hamid, The Angeles Clinic and Research Institute, Los Angeles, CA; Rene Gonzalez, Karl Lewis, University of Colorado; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver, CO; Jeffrey A. Sosman, Igor Puzanov, Vanderbilt University Medical Center, Nashville, TN; Lynn Schuchter, University of Pennsylvania Abramson Cancer Center; Nageatte Ibrahim, Elizabeth Cunningham, Merck; Peng Sun, Amy S. Kline, Heather Del Buono, Diane Opatt McDowell, GlaxoSmithKline, Philadelphia, PA; Donald Lawrence and Kiran Patel, Incyte Corporation, Wilmington, DE; and Keith T. Flaherty, Massachusetts General Hospital Cancer Center, Boston, MA.
| | - Jeffrey S Weber
- Georgina V. Long, Melanoma Institute Australia; The University of Sydney; Richard F. Kefford, Melanoma Institute Australia; The University of Sydney; Macquarie University, Sydney; Westmead Hospital, Westmead; Jonathan Cebon, Austin Health, Melbourne, Victoria, Australia; Jeffrey S. Weber and Ragini Kudchadkar, Moffitt Cancer Center, Tampa, FL; Jeffrey R. Infante and Howard A. Burris III, Sarah Cannon Research Institute/Tennessee Oncology; Kevin B. Kim, California Pacific Medical Center; Adil Daud, Alain Algazi, University of California, San Francisco, San Francisco; Omid Hamid, The Angeles Clinic and Research Institute, Los Angeles, CA; Rene Gonzalez, Karl Lewis, University of Colorado; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver, CO; Jeffrey A. Sosman, Igor Puzanov, Vanderbilt University Medical Center, Nashville, TN; Lynn Schuchter, University of Pennsylvania Abramson Cancer Center; Nageatte Ibrahim, Elizabeth Cunningham, Merck; Peng Sun, Amy S. Kline, Heather Del Buono, Diane Opatt McDowell, GlaxoSmithKline, Philadelphia, PA; Donald Lawrence and Kiran Patel, Incyte Corporation, Wilmington, DE; and Keith T. Flaherty, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Jeffrey R Infante
- Georgina V. Long, Melanoma Institute Australia; The University of Sydney; Richard F. Kefford, Melanoma Institute Australia; The University of Sydney; Macquarie University, Sydney; Westmead Hospital, Westmead; Jonathan Cebon, Austin Health, Melbourne, Victoria, Australia; Jeffrey S. Weber and Ragini Kudchadkar, Moffitt Cancer Center, Tampa, FL; Jeffrey R. Infante and Howard A. Burris III, Sarah Cannon Research Institute/Tennessee Oncology; Kevin B. Kim, California Pacific Medical Center; Adil Daud, Alain Algazi, University of California, San Francisco, San Francisco; Omid Hamid, The Angeles Clinic and Research Institute, Los Angeles, CA; Rene Gonzalez, Karl Lewis, University of Colorado; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver, CO; Jeffrey A. Sosman, Igor Puzanov, Vanderbilt University Medical Center, Nashville, TN; Lynn Schuchter, University of Pennsylvania Abramson Cancer Center; Nageatte Ibrahim, Elizabeth Cunningham, Merck; Peng Sun, Amy S. Kline, Heather Del Buono, Diane Opatt McDowell, GlaxoSmithKline, Philadelphia, PA; Donald Lawrence and Kiran Patel, Incyte Corporation, Wilmington, DE; and Keith T. Flaherty, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Kevin B Kim
- Georgina V. Long, Melanoma Institute Australia; The University of Sydney; Richard F. Kefford, Melanoma Institute Australia; The University of Sydney; Macquarie University, Sydney; Westmead Hospital, Westmead; Jonathan Cebon, Austin Health, Melbourne, Victoria, Australia; Jeffrey S. Weber and Ragini Kudchadkar, Moffitt Cancer Center, Tampa, FL; Jeffrey R. Infante and Howard A. Burris III, Sarah Cannon Research Institute/Tennessee Oncology; Kevin B. Kim, California Pacific Medical Center; Adil Daud, Alain Algazi, University of California, San Francisco, San Francisco; Omid Hamid, The Angeles Clinic and Research Institute, Los Angeles, CA; Rene Gonzalez, Karl Lewis, University of Colorado; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver, CO; Jeffrey A. Sosman, Igor Puzanov, Vanderbilt University Medical Center, Nashville, TN; Lynn Schuchter, University of Pennsylvania Abramson Cancer Center; Nageatte Ibrahim, Elizabeth Cunningham, Merck; Peng Sun, Amy S. Kline, Heather Del Buono, Diane Opatt McDowell, GlaxoSmithKline, Philadelphia, PA; Donald Lawrence and Kiran Patel, Incyte Corporation, Wilmington, DE; and Keith T. Flaherty, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Adil Daud
- Georgina V. Long, Melanoma Institute Australia; The University of Sydney; Richard F. Kefford, Melanoma Institute Australia; The University of Sydney; Macquarie University, Sydney; Westmead Hospital, Westmead; Jonathan Cebon, Austin Health, Melbourne, Victoria, Australia; Jeffrey S. Weber and Ragini Kudchadkar, Moffitt Cancer Center, Tampa, FL; Jeffrey R. Infante and Howard A. Burris III, Sarah Cannon Research Institute/Tennessee Oncology; Kevin B. Kim, California Pacific Medical Center; Adil Daud, Alain Algazi, University of California, San Francisco, San Francisco; Omid Hamid, The Angeles Clinic and Research Institute, Los Angeles, CA; Rene Gonzalez, Karl Lewis, University of Colorado; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver, CO; Jeffrey A. Sosman, Igor Puzanov, Vanderbilt University Medical Center, Nashville, TN; Lynn Schuchter, University of Pennsylvania Abramson Cancer Center; Nageatte Ibrahim, Elizabeth Cunningham, Merck; Peng Sun, Amy S. Kline, Heather Del Buono, Diane Opatt McDowell, GlaxoSmithKline, Philadelphia, PA; Donald Lawrence and Kiran Patel, Incyte Corporation, Wilmington, DE; and Keith T. Flaherty, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Rene Gonzalez
- Georgina V. Long, Melanoma Institute Australia; The University of Sydney; Richard F. Kefford, Melanoma Institute Australia; The University of Sydney; Macquarie University, Sydney; Westmead Hospital, Westmead; Jonathan Cebon, Austin Health, Melbourne, Victoria, Australia; Jeffrey S. Weber and Ragini Kudchadkar, Moffitt Cancer Center, Tampa, FL; Jeffrey R. Infante and Howard A. Burris III, Sarah Cannon Research Institute/Tennessee Oncology; Kevin B. Kim, California Pacific Medical Center; Adil Daud, Alain Algazi, University of California, San Francisco, San Francisco; Omid Hamid, The Angeles Clinic and Research Institute, Los Angeles, CA; Rene Gonzalez, Karl Lewis, University of Colorado; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver, CO; Jeffrey A. Sosman, Igor Puzanov, Vanderbilt University Medical Center, Nashville, TN; Lynn Schuchter, University of Pennsylvania Abramson Cancer Center; Nageatte Ibrahim, Elizabeth Cunningham, Merck; Peng Sun, Amy S. Kline, Heather Del Buono, Diane Opatt McDowell, GlaxoSmithKline, Philadelphia, PA; Donald Lawrence and Kiran Patel, Incyte Corporation, Wilmington, DE; and Keith T. Flaherty, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Jeffrey A Sosman
- Georgina V. Long, Melanoma Institute Australia; The University of Sydney; Richard F. Kefford, Melanoma Institute Australia; The University of Sydney; Macquarie University, Sydney; Westmead Hospital, Westmead; Jonathan Cebon, Austin Health, Melbourne, Victoria, Australia; Jeffrey S. Weber and Ragini Kudchadkar, Moffitt Cancer Center, Tampa, FL; Jeffrey R. Infante and Howard A. Burris III, Sarah Cannon Research Institute/Tennessee Oncology; Kevin B. Kim, California Pacific Medical Center; Adil Daud, Alain Algazi, University of California, San Francisco, San Francisco; Omid Hamid, The Angeles Clinic and Research Institute, Los Angeles, CA; Rene Gonzalez, Karl Lewis, University of Colorado; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver, CO; Jeffrey A. Sosman, Igor Puzanov, Vanderbilt University Medical Center, Nashville, TN; Lynn Schuchter, University of Pennsylvania Abramson Cancer Center; Nageatte Ibrahim, Elizabeth Cunningham, Merck; Peng Sun, Amy S. Kline, Heather Del Buono, Diane Opatt McDowell, GlaxoSmithKline, Philadelphia, PA; Donald Lawrence and Kiran Patel, Incyte Corporation, Wilmington, DE; and Keith T. Flaherty, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Omid Hamid
- Georgina V. Long, Melanoma Institute Australia; The University of Sydney; Richard F. Kefford, Melanoma Institute Australia; The University of Sydney; Macquarie University, Sydney; Westmead Hospital, Westmead; Jonathan Cebon, Austin Health, Melbourne, Victoria, Australia; Jeffrey S. Weber and Ragini Kudchadkar, Moffitt Cancer Center, Tampa, FL; Jeffrey R. Infante and Howard A. Burris III, Sarah Cannon Research Institute/Tennessee Oncology; Kevin B. Kim, California Pacific Medical Center; Adil Daud, Alain Algazi, University of California, San Francisco, San Francisco; Omid Hamid, The Angeles Clinic and Research Institute, Los Angeles, CA; Rene Gonzalez, Karl Lewis, University of Colorado; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver, CO; Jeffrey A. Sosman, Igor Puzanov, Vanderbilt University Medical Center, Nashville, TN; Lynn Schuchter, University of Pennsylvania Abramson Cancer Center; Nageatte Ibrahim, Elizabeth Cunningham, Merck; Peng Sun, Amy S. Kline, Heather Del Buono, Diane Opatt McDowell, GlaxoSmithKline, Philadelphia, PA; Donald Lawrence and Kiran Patel, Incyte Corporation, Wilmington, DE; and Keith T. Flaherty, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Lynn Schuchter
- Georgina V. Long, Melanoma Institute Australia; The University of Sydney; Richard F. Kefford, Melanoma Institute Australia; The University of Sydney; Macquarie University, Sydney; Westmead Hospital, Westmead; Jonathan Cebon, Austin Health, Melbourne, Victoria, Australia; Jeffrey S. Weber and Ragini Kudchadkar, Moffitt Cancer Center, Tampa, FL; Jeffrey R. Infante and Howard A. Burris III, Sarah Cannon Research Institute/Tennessee Oncology; Kevin B. Kim, California Pacific Medical Center; Adil Daud, Alain Algazi, University of California, San Francisco, San Francisco; Omid Hamid, The Angeles Clinic and Research Institute, Los Angeles, CA; Rene Gonzalez, Karl Lewis, University of Colorado; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver, CO; Jeffrey A. Sosman, Igor Puzanov, Vanderbilt University Medical Center, Nashville, TN; Lynn Schuchter, University of Pennsylvania Abramson Cancer Center; Nageatte Ibrahim, Elizabeth Cunningham, Merck; Peng Sun, Amy S. Kline, Heather Del Buono, Diane Opatt McDowell, GlaxoSmithKline, Philadelphia, PA; Donald Lawrence and Kiran Patel, Incyte Corporation, Wilmington, DE; and Keith T. Flaherty, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Jonathan Cebon
- Georgina V. Long, Melanoma Institute Australia; The University of Sydney; Richard F. Kefford, Melanoma Institute Australia; The University of Sydney; Macquarie University, Sydney; Westmead Hospital, Westmead; Jonathan Cebon, Austin Health, Melbourne, Victoria, Australia; Jeffrey S. Weber and Ragini Kudchadkar, Moffitt Cancer Center, Tampa, FL; Jeffrey R. Infante and Howard A. Burris III, Sarah Cannon Research Institute/Tennessee Oncology; Kevin B. Kim, California Pacific Medical Center; Adil Daud, Alain Algazi, University of California, San Francisco, San Francisco; Omid Hamid, The Angeles Clinic and Research Institute, Los Angeles, CA; Rene Gonzalez, Karl Lewis, University of Colorado; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver, CO; Jeffrey A. Sosman, Igor Puzanov, Vanderbilt University Medical Center, Nashville, TN; Lynn Schuchter, University of Pennsylvania Abramson Cancer Center; Nageatte Ibrahim, Elizabeth Cunningham, Merck; Peng Sun, Amy S. Kline, Heather Del Buono, Diane Opatt McDowell, GlaxoSmithKline, Philadelphia, PA; Donald Lawrence and Kiran Patel, Incyte Corporation, Wilmington, DE; and Keith T. Flaherty, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Richard F Kefford
- Georgina V. Long, Melanoma Institute Australia; The University of Sydney; Richard F. Kefford, Melanoma Institute Australia; The University of Sydney; Macquarie University, Sydney; Westmead Hospital, Westmead; Jonathan Cebon, Austin Health, Melbourne, Victoria, Australia; Jeffrey S. Weber and Ragini Kudchadkar, Moffitt Cancer Center, Tampa, FL; Jeffrey R. Infante and Howard A. Burris III, Sarah Cannon Research Institute/Tennessee Oncology; Kevin B. Kim, California Pacific Medical Center; Adil Daud, Alain Algazi, University of California, San Francisco, San Francisco; Omid Hamid, The Angeles Clinic and Research Institute, Los Angeles, CA; Rene Gonzalez, Karl Lewis, University of Colorado; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver, CO; Jeffrey A. Sosman, Igor Puzanov, Vanderbilt University Medical Center, Nashville, TN; Lynn Schuchter, University of Pennsylvania Abramson Cancer Center; Nageatte Ibrahim, Elizabeth Cunningham, Merck; Peng Sun, Amy S. Kline, Heather Del Buono, Diane Opatt McDowell, GlaxoSmithKline, Philadelphia, PA; Donald Lawrence and Kiran Patel, Incyte Corporation, Wilmington, DE; and Keith T. Flaherty, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Donald Lawrence
- Georgina V. Long, Melanoma Institute Australia; The University of Sydney; Richard F. Kefford, Melanoma Institute Australia; The University of Sydney; Macquarie University, Sydney; Westmead Hospital, Westmead; Jonathan Cebon, Austin Health, Melbourne, Victoria, Australia; Jeffrey S. Weber and Ragini Kudchadkar, Moffitt Cancer Center, Tampa, FL; Jeffrey R. Infante and Howard A. Burris III, Sarah Cannon Research Institute/Tennessee Oncology; Kevin B. Kim, California Pacific Medical Center; Adil Daud, Alain Algazi, University of California, San Francisco, San Francisco; Omid Hamid, The Angeles Clinic and Research Institute, Los Angeles, CA; Rene Gonzalez, Karl Lewis, University of Colorado; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver, CO; Jeffrey A. Sosman, Igor Puzanov, Vanderbilt University Medical Center, Nashville, TN; Lynn Schuchter, University of Pennsylvania Abramson Cancer Center; Nageatte Ibrahim, Elizabeth Cunningham, Merck; Peng Sun, Amy S. Kline, Heather Del Buono, Diane Opatt McDowell, GlaxoSmithKline, Philadelphia, PA; Donald Lawrence and Kiran Patel, Incyte Corporation, Wilmington, DE; and Keith T. Flaherty, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Ragini Kudchadkar
- Georgina V. Long, Melanoma Institute Australia; The University of Sydney; Richard F. Kefford, Melanoma Institute Australia; The University of Sydney; Macquarie University, Sydney; Westmead Hospital, Westmead; Jonathan Cebon, Austin Health, Melbourne, Victoria, Australia; Jeffrey S. Weber and Ragini Kudchadkar, Moffitt Cancer Center, Tampa, FL; Jeffrey R. Infante and Howard A. Burris III, Sarah Cannon Research Institute/Tennessee Oncology; Kevin B. Kim, California Pacific Medical Center; Adil Daud, Alain Algazi, University of California, San Francisco, San Francisco; Omid Hamid, The Angeles Clinic and Research Institute, Los Angeles, CA; Rene Gonzalez, Karl Lewis, University of Colorado; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver, CO; Jeffrey A. Sosman, Igor Puzanov, Vanderbilt University Medical Center, Nashville, TN; Lynn Schuchter, University of Pennsylvania Abramson Cancer Center; Nageatte Ibrahim, Elizabeth Cunningham, Merck; Peng Sun, Amy S. Kline, Heather Del Buono, Diane Opatt McDowell, GlaxoSmithKline, Philadelphia, PA; Donald Lawrence and Kiran Patel, Incyte Corporation, Wilmington, DE; and Keith T. Flaherty, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Howard A Burris
- Georgina V. Long, Melanoma Institute Australia; The University of Sydney; Richard F. Kefford, Melanoma Institute Australia; The University of Sydney; Macquarie University, Sydney; Westmead Hospital, Westmead; Jonathan Cebon, Austin Health, Melbourne, Victoria, Australia; Jeffrey S. Weber and Ragini Kudchadkar, Moffitt Cancer Center, Tampa, FL; Jeffrey R. Infante and Howard A. Burris III, Sarah Cannon Research Institute/Tennessee Oncology; Kevin B. Kim, California Pacific Medical Center; Adil Daud, Alain Algazi, University of California, San Francisco, San Francisco; Omid Hamid, The Angeles Clinic and Research Institute, Los Angeles, CA; Rene Gonzalez, Karl Lewis, University of Colorado; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver, CO; Jeffrey A. Sosman, Igor Puzanov, Vanderbilt University Medical Center, Nashville, TN; Lynn Schuchter, University of Pennsylvania Abramson Cancer Center; Nageatte Ibrahim, Elizabeth Cunningham, Merck; Peng Sun, Amy S. Kline, Heather Del Buono, Diane Opatt McDowell, GlaxoSmithKline, Philadelphia, PA; Donald Lawrence and Kiran Patel, Incyte Corporation, Wilmington, DE; and Keith T. Flaherty, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Gerald S Falchook
- Georgina V. Long, Melanoma Institute Australia; The University of Sydney; Richard F. Kefford, Melanoma Institute Australia; The University of Sydney; Macquarie University, Sydney; Westmead Hospital, Westmead; Jonathan Cebon, Austin Health, Melbourne, Victoria, Australia; Jeffrey S. Weber and Ragini Kudchadkar, Moffitt Cancer Center, Tampa, FL; Jeffrey R. Infante and Howard A. Burris III, Sarah Cannon Research Institute/Tennessee Oncology; Kevin B. Kim, California Pacific Medical Center; Adil Daud, Alain Algazi, University of California, San Francisco, San Francisco; Omid Hamid, The Angeles Clinic and Research Institute, Los Angeles, CA; Rene Gonzalez, Karl Lewis, University of Colorado; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver, CO; Jeffrey A. Sosman, Igor Puzanov, Vanderbilt University Medical Center, Nashville, TN; Lynn Schuchter, University of Pennsylvania Abramson Cancer Center; Nageatte Ibrahim, Elizabeth Cunningham, Merck; Peng Sun, Amy S. Kline, Heather Del Buono, Diane Opatt McDowell, GlaxoSmithKline, Philadelphia, PA; Donald Lawrence and Kiran Patel, Incyte Corporation, Wilmington, DE; and Keith T. Flaherty, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Alain Algazi
- Georgina V. Long, Melanoma Institute Australia; The University of Sydney; Richard F. Kefford, Melanoma Institute Australia; The University of Sydney; Macquarie University, Sydney; Westmead Hospital, Westmead; Jonathan Cebon, Austin Health, Melbourne, Victoria, Australia; Jeffrey S. Weber and Ragini Kudchadkar, Moffitt Cancer Center, Tampa, FL; Jeffrey R. Infante and Howard A. Burris III, Sarah Cannon Research Institute/Tennessee Oncology; Kevin B. Kim, California Pacific Medical Center; Adil Daud, Alain Algazi, University of California, San Francisco, San Francisco; Omid Hamid, The Angeles Clinic and Research Institute, Los Angeles, CA; Rene Gonzalez, Karl Lewis, University of Colorado; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver, CO; Jeffrey A. Sosman, Igor Puzanov, Vanderbilt University Medical Center, Nashville, TN; Lynn Schuchter, University of Pennsylvania Abramson Cancer Center; Nageatte Ibrahim, Elizabeth Cunningham, Merck; Peng Sun, Amy S. Kline, Heather Del Buono, Diane Opatt McDowell, GlaxoSmithKline, Philadelphia, PA; Donald Lawrence and Kiran Patel, Incyte Corporation, Wilmington, DE; and Keith T. Flaherty, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Karl Lewis
- Georgina V. Long, Melanoma Institute Australia; The University of Sydney; Richard F. Kefford, Melanoma Institute Australia; The University of Sydney; Macquarie University, Sydney; Westmead Hospital, Westmead; Jonathan Cebon, Austin Health, Melbourne, Victoria, Australia; Jeffrey S. Weber and Ragini Kudchadkar, Moffitt Cancer Center, Tampa, FL; Jeffrey R. Infante and Howard A. Burris III, Sarah Cannon Research Institute/Tennessee Oncology; Kevin B. Kim, California Pacific Medical Center; Adil Daud, Alain Algazi, University of California, San Francisco, San Francisco; Omid Hamid, The Angeles Clinic and Research Institute, Los Angeles, CA; Rene Gonzalez, Karl Lewis, University of Colorado; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver, CO; Jeffrey A. Sosman, Igor Puzanov, Vanderbilt University Medical Center, Nashville, TN; Lynn Schuchter, University of Pennsylvania Abramson Cancer Center; Nageatte Ibrahim, Elizabeth Cunningham, Merck; Peng Sun, Amy S. Kline, Heather Del Buono, Diane Opatt McDowell, GlaxoSmithKline, Philadelphia, PA; Donald Lawrence and Kiran Patel, Incyte Corporation, Wilmington, DE; and Keith T. Flaherty, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Igor Puzanov
- Georgina V. Long, Melanoma Institute Australia; The University of Sydney; Richard F. Kefford, Melanoma Institute Australia; The University of Sydney; Macquarie University, Sydney; Westmead Hospital, Westmead; Jonathan Cebon, Austin Health, Melbourne, Victoria, Australia; Jeffrey S. Weber and Ragini Kudchadkar, Moffitt Cancer Center, Tampa, FL; Jeffrey R. Infante and Howard A. Burris III, Sarah Cannon Research Institute/Tennessee Oncology; Kevin B. Kim, California Pacific Medical Center; Adil Daud, Alain Algazi, University of California, San Francisco, San Francisco; Omid Hamid, The Angeles Clinic and Research Institute, Los Angeles, CA; Rene Gonzalez, Karl Lewis, University of Colorado; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver, CO; Jeffrey A. Sosman, Igor Puzanov, Vanderbilt University Medical Center, Nashville, TN; Lynn Schuchter, University of Pennsylvania Abramson Cancer Center; Nageatte Ibrahim, Elizabeth Cunningham, Merck; Peng Sun, Amy S. Kline, Heather Del Buono, Diane Opatt McDowell, GlaxoSmithKline, Philadelphia, PA; Donald Lawrence and Kiran Patel, Incyte Corporation, Wilmington, DE; and Keith T. Flaherty, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Nageatte Ibrahim
- Georgina V. Long, Melanoma Institute Australia; The University of Sydney; Richard F. Kefford, Melanoma Institute Australia; The University of Sydney; Macquarie University, Sydney; Westmead Hospital, Westmead; Jonathan Cebon, Austin Health, Melbourne, Victoria, Australia; Jeffrey S. Weber and Ragini Kudchadkar, Moffitt Cancer Center, Tampa, FL; Jeffrey R. Infante and Howard A. Burris III, Sarah Cannon Research Institute/Tennessee Oncology; Kevin B. Kim, California Pacific Medical Center; Adil Daud, Alain Algazi, University of California, San Francisco, San Francisco; Omid Hamid, The Angeles Clinic and Research Institute, Los Angeles, CA; Rene Gonzalez, Karl Lewis, University of Colorado; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver, CO; Jeffrey A. Sosman, Igor Puzanov, Vanderbilt University Medical Center, Nashville, TN; Lynn Schuchter, University of Pennsylvania Abramson Cancer Center; Nageatte Ibrahim, Elizabeth Cunningham, Merck; Peng Sun, Amy S. Kline, Heather Del Buono, Diane Opatt McDowell, GlaxoSmithKline, Philadelphia, PA; Donald Lawrence and Kiran Patel, Incyte Corporation, Wilmington, DE; and Keith T. Flaherty, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Peng Sun
- Georgina V. Long, Melanoma Institute Australia; The University of Sydney; Richard F. Kefford, Melanoma Institute Australia; The University of Sydney; Macquarie University, Sydney; Westmead Hospital, Westmead; Jonathan Cebon, Austin Health, Melbourne, Victoria, Australia; Jeffrey S. Weber and Ragini Kudchadkar, Moffitt Cancer Center, Tampa, FL; Jeffrey R. Infante and Howard A. Burris III, Sarah Cannon Research Institute/Tennessee Oncology; Kevin B. Kim, California Pacific Medical Center; Adil Daud, Alain Algazi, University of California, San Francisco, San Francisco; Omid Hamid, The Angeles Clinic and Research Institute, Los Angeles, CA; Rene Gonzalez, Karl Lewis, University of Colorado; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver, CO; Jeffrey A. Sosman, Igor Puzanov, Vanderbilt University Medical Center, Nashville, TN; Lynn Schuchter, University of Pennsylvania Abramson Cancer Center; Nageatte Ibrahim, Elizabeth Cunningham, Merck; Peng Sun, Amy S. Kline, Heather Del Buono, Diane Opatt McDowell, GlaxoSmithKline, Philadelphia, PA; Donald Lawrence and Kiran Patel, Incyte Corporation, Wilmington, DE; and Keith T. Flaherty, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Elizabeth Cunningham
- Georgina V. Long, Melanoma Institute Australia; The University of Sydney; Richard F. Kefford, Melanoma Institute Australia; The University of Sydney; Macquarie University, Sydney; Westmead Hospital, Westmead; Jonathan Cebon, Austin Health, Melbourne, Victoria, Australia; Jeffrey S. Weber and Ragini Kudchadkar, Moffitt Cancer Center, Tampa, FL; Jeffrey R. Infante and Howard A. Burris III, Sarah Cannon Research Institute/Tennessee Oncology; Kevin B. Kim, California Pacific Medical Center; Adil Daud, Alain Algazi, University of California, San Francisco, San Francisco; Omid Hamid, The Angeles Clinic and Research Institute, Los Angeles, CA; Rene Gonzalez, Karl Lewis, University of Colorado; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver, CO; Jeffrey A. Sosman, Igor Puzanov, Vanderbilt University Medical Center, Nashville, TN; Lynn Schuchter, University of Pennsylvania Abramson Cancer Center; Nageatte Ibrahim, Elizabeth Cunningham, Merck; Peng Sun, Amy S. Kline, Heather Del Buono, Diane Opatt McDowell, GlaxoSmithKline, Philadelphia, PA; Donald Lawrence and Kiran Patel, Incyte Corporation, Wilmington, DE; and Keith T. Flaherty, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Amy S Kline
- Georgina V. Long, Melanoma Institute Australia; The University of Sydney; Richard F. Kefford, Melanoma Institute Australia; The University of Sydney; Macquarie University, Sydney; Westmead Hospital, Westmead; Jonathan Cebon, Austin Health, Melbourne, Victoria, Australia; Jeffrey S. Weber and Ragini Kudchadkar, Moffitt Cancer Center, Tampa, FL; Jeffrey R. Infante and Howard A. Burris III, Sarah Cannon Research Institute/Tennessee Oncology; Kevin B. Kim, California Pacific Medical Center; Adil Daud, Alain Algazi, University of California, San Francisco, San Francisco; Omid Hamid, The Angeles Clinic and Research Institute, Los Angeles, CA; Rene Gonzalez, Karl Lewis, University of Colorado; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver, CO; Jeffrey A. Sosman, Igor Puzanov, Vanderbilt University Medical Center, Nashville, TN; Lynn Schuchter, University of Pennsylvania Abramson Cancer Center; Nageatte Ibrahim, Elizabeth Cunningham, Merck; Peng Sun, Amy S. Kline, Heather Del Buono, Diane Opatt McDowell, GlaxoSmithKline, Philadelphia, PA; Donald Lawrence and Kiran Patel, Incyte Corporation, Wilmington, DE; and Keith T. Flaherty, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Heather Del Buono
- Georgina V. Long, Melanoma Institute Australia; The University of Sydney; Richard F. Kefford, Melanoma Institute Australia; The University of Sydney; Macquarie University, Sydney; Westmead Hospital, Westmead; Jonathan Cebon, Austin Health, Melbourne, Victoria, Australia; Jeffrey S. Weber and Ragini Kudchadkar, Moffitt Cancer Center, Tampa, FL; Jeffrey R. Infante and Howard A. Burris III, Sarah Cannon Research Institute/Tennessee Oncology; Kevin B. Kim, California Pacific Medical Center; Adil Daud, Alain Algazi, University of California, San Francisco, San Francisco; Omid Hamid, The Angeles Clinic and Research Institute, Los Angeles, CA; Rene Gonzalez, Karl Lewis, University of Colorado; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver, CO; Jeffrey A. Sosman, Igor Puzanov, Vanderbilt University Medical Center, Nashville, TN; Lynn Schuchter, University of Pennsylvania Abramson Cancer Center; Nageatte Ibrahim, Elizabeth Cunningham, Merck; Peng Sun, Amy S. Kline, Heather Del Buono, Diane Opatt McDowell, GlaxoSmithKline, Philadelphia, PA; Donald Lawrence and Kiran Patel, Incyte Corporation, Wilmington, DE; and Keith T. Flaherty, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Diane Opatt McDowell
- Georgina V. Long, Melanoma Institute Australia; The University of Sydney; Richard F. Kefford, Melanoma Institute Australia; The University of Sydney; Macquarie University, Sydney; Westmead Hospital, Westmead; Jonathan Cebon, Austin Health, Melbourne, Victoria, Australia; Jeffrey S. Weber and Ragini Kudchadkar, Moffitt Cancer Center, Tampa, FL; Jeffrey R. Infante and Howard A. Burris III, Sarah Cannon Research Institute/Tennessee Oncology; Kevin B. Kim, California Pacific Medical Center; Adil Daud, Alain Algazi, University of California, San Francisco, San Francisco; Omid Hamid, The Angeles Clinic and Research Institute, Los Angeles, CA; Rene Gonzalez, Karl Lewis, University of Colorado; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver, CO; Jeffrey A. Sosman, Igor Puzanov, Vanderbilt University Medical Center, Nashville, TN; Lynn Schuchter, University of Pennsylvania Abramson Cancer Center; Nageatte Ibrahim, Elizabeth Cunningham, Merck; Peng Sun, Amy S. Kline, Heather Del Buono, Diane Opatt McDowell, GlaxoSmithKline, Philadelphia, PA; Donald Lawrence and Kiran Patel, Incyte Corporation, Wilmington, DE; and Keith T. Flaherty, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Kiran Patel
- Georgina V. Long, Melanoma Institute Australia; The University of Sydney; Richard F. Kefford, Melanoma Institute Australia; The University of Sydney; Macquarie University, Sydney; Westmead Hospital, Westmead; Jonathan Cebon, Austin Health, Melbourne, Victoria, Australia; Jeffrey S. Weber and Ragini Kudchadkar, Moffitt Cancer Center, Tampa, FL; Jeffrey R. Infante and Howard A. Burris III, Sarah Cannon Research Institute/Tennessee Oncology; Kevin B. Kim, California Pacific Medical Center; Adil Daud, Alain Algazi, University of California, San Francisco, San Francisco; Omid Hamid, The Angeles Clinic and Research Institute, Los Angeles, CA; Rene Gonzalez, Karl Lewis, University of Colorado; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver, CO; Jeffrey A. Sosman, Igor Puzanov, Vanderbilt University Medical Center, Nashville, TN; Lynn Schuchter, University of Pennsylvania Abramson Cancer Center; Nageatte Ibrahim, Elizabeth Cunningham, Merck; Peng Sun, Amy S. Kline, Heather Del Buono, Diane Opatt McDowell, GlaxoSmithKline, Philadelphia, PA; Donald Lawrence and Kiran Patel, Incyte Corporation, Wilmington, DE; and Keith T. Flaherty, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Keith T Flaherty
- Georgina V. Long, Melanoma Institute Australia; The University of Sydney; Richard F. Kefford, Melanoma Institute Australia; The University of Sydney; Macquarie University, Sydney; Westmead Hospital, Westmead; Jonathan Cebon, Austin Health, Melbourne, Victoria, Australia; Jeffrey S. Weber and Ragini Kudchadkar, Moffitt Cancer Center, Tampa, FL; Jeffrey R. Infante and Howard A. Burris III, Sarah Cannon Research Institute/Tennessee Oncology; Kevin B. Kim, California Pacific Medical Center; Adil Daud, Alain Algazi, University of California, San Francisco, San Francisco; Omid Hamid, The Angeles Clinic and Research Institute, Los Angeles, CA; Rene Gonzalez, Karl Lewis, University of Colorado; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver, CO; Jeffrey A. Sosman, Igor Puzanov, Vanderbilt University Medical Center, Nashville, TN; Lynn Schuchter, University of Pennsylvania Abramson Cancer Center; Nageatte Ibrahim, Elizabeth Cunningham, Merck; Peng Sun, Amy S. Kline, Heather Del Buono, Diane Opatt McDowell, GlaxoSmithKline, Philadelphia, PA; Donald Lawrence and Kiran Patel, Incyte Corporation, Wilmington, DE; and Keith T. Flaherty, Massachusetts General Hospital Cancer Center, Boston, MA
| |
Collapse
|
6009
|
Meyer N. [What's new in oncodermatology in 2015?]. Ann Dermatol Venereol 2016; 142 Suppl 3:S36-48. [PMID: 26792413 DOI: 10.1016/s0151-9638(16)30005-9] [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: 11/28/2022]
Abstract
Oncodermatology is the topic of constant innovation since 5 years. These innovations have drammatically modified the prognostic of skin cancers and induced a change in the paradigm that drives the whole oncology. Once again in 2015, melanoma is the key topic of scientific communication, with promising results. However, unmet clinical needs must be kept in light. Some diseases, such as squamous cell carcinoma or cutaneous lymphoma remain poorly evaluated. This article was designed as a review of litterature. All keywords of oncodermatology were searched in the main journals of dermatology, oncology and internal medicine. Despite methodologic restrictions, the number of articles imposed a choice based on opinion. The final goal was to transmit author's enthousiasm and the most important results.
Collapse
Affiliation(s)
- N Meyer
- Dermatologie, université Paul-Sabatier - Toulouse III et Institut Universitaire du Cancer de Toulouse, 1, rue Irène-Joliot- Curie, 31059 Toulouse Cedex 9 ; INSERM UMR 1037, CRCT, Toulouse, France.
| |
Collapse
|
6010
|
Spranger S, Sivan A, Corrales L, Gajewski TF. Tumor and Host Factors Controlling Antitumor Immunity and Efficacy of Cancer Immunotherapy. Adv Immunol 2016; 130:75-93. [PMID: 26923000 DOI: 10.1016/bs.ai.2015.12.003] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Despite recent clinical advances in immunotherapy, a fraction of cancer patients fails to respond to these interventions. Evidence from preclinical mouse models as well as clinical samples has provided evidence that the extent of activated T cell infiltration within the tumor microenvironment is associated with clinical response to immunotherapies including checkpoint blockade. Therefore, understanding the molecular mechanisms mediating the lack of T cell infiltration into the tumor microenvironment will be instrumental for the development of new therapeutic strategies to render those patients immunotherapy responsive. Recent data have suggested that major sources of intersubject heterogeneity include differences in somatic mutations in specific oncogene pathways between cancers of individual subjects and also environmental factors including commensal microbial composition. Successful identification of such causal factors should lead to new therapeutic approaches that may facilitate T cell entry into noninflamed tumors and expand the fraction of patients capable of responding to novel immunotherapies.
Collapse
Affiliation(s)
| | - Ayelet Sivan
- The University of Chicago, Chicago, IL, United States
| | | | | |
Collapse
|
6011
|
Viteri S, González-Cao M, Barrón F, Riso A, Rosell R. Results of clinical trials with anti-programmed death 1/programmed death ligand 1 inhibitors in lung cancer. Transl Lung Cancer Res 2016; 4:756-62. [PMID: 26798585 DOI: 10.3978/j.issn.2218-6751.2015.12.06] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
One of the main hallmarks of cancer is the capability of evading immune destruction. In order to drive tumor progression, malignant cells are able to promote immunosuppressive mechanisms avoiding recognition and elimination. Increasing knowledge of the mechanisms of immune tolerance has led to the identification of several membrane receptors strongly implicated in this cancer feature: the immune checkpoints. Among them, programmed death 1 (PD-1) receptors and their ligands have been identified as potential targets for a new anti-cancer therapeutic approach: the use of immune-modulatory monoclonal antibodies designed to interrupt the immune escape activated by the interaction of PD-1 receptors and their ligands. Five of these antibodies are now in their late stages of clinical development and this review will summarize their up-to-date efficacy and toxicity data.
Collapse
Affiliation(s)
- Santiago Viteri
- 1 Instituto Oncológico Dr Rosell, Quirón-Dexeus University Hospital, Barcelona, Spain ; 2 Instituto Nacional de Cancerología, México City, México
| | - María González-Cao
- 1 Instituto Oncológico Dr Rosell, Quirón-Dexeus University Hospital, Barcelona, Spain ; 2 Instituto Nacional de Cancerología, México City, México
| | - Feliciano Barrón
- 1 Instituto Oncológico Dr Rosell, Quirón-Dexeus University Hospital, Barcelona, Spain ; 2 Instituto Nacional de Cancerología, México City, México
| | - Aldo Riso
- 1 Instituto Oncológico Dr Rosell, Quirón-Dexeus University Hospital, Barcelona, Spain ; 2 Instituto Nacional de Cancerología, México City, México
| | - Rafael Rosell
- 1 Instituto Oncológico Dr Rosell, Quirón-Dexeus University Hospital, Barcelona, Spain ; 2 Instituto Nacional de Cancerología, México City, México
| |
Collapse
|
6012
|
Yuan J, Hegde PS, Clynes R, Foukas PG, Harari A, Kleen TO, Kvistborg P, Maccalli C, Maecker HT, Page DB, Robins H, Song W, Stack EC, Wang E, Whiteside TL, Zhao Y, Zwierzina H, Butterfield LH, Fox BA. Novel technologies and emerging biomarkers for personalized cancer immunotherapy. J Immunother Cancer 2016. [PMID: 26788324 DOI: 10.1186/s40425-016-0107-3.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
The culmination of over a century's work to understand the role of the immune system in tumor control has led to the recent advances in cancer immunotherapies that have resulted in durable clinical responses in patients with a variety of malignancies. Cancer immunotherapies are rapidly changing traditional treatment paradigms and expanding the therapeutic landscape for cancer patients. However, despite the current success of these therapies, not all patients respond to immunotherapy and even those that do often experience toxicities. Thus, there is a growing need to identify predictive and prognostic biomarkers that enhance our understanding of the mechanisms underlying the complex interactions between the immune system and cancer. Therefore, the Society for Immunotherapy of Cancer (SITC) reconvened an Immune Biomarkers Task Force to review state of the art technologies, identify current hurdlers, and make recommendations for the field. As a product of this task force, Working Group 2 (WG2), consisting of international experts from academia and industry, assembled to identify and discuss promising technologies for biomarker discovery and validation. Thus, this WG2 consensus paper will focus on the current status of emerging biomarkers for immune checkpoint blockade therapy and discuss novel technologies as well as high dimensional data analysis platforms that will be pivotal for future biomarker research. In addition, this paper will include a brief overview of the current challenges with recommendations for future biomarker discovery.
Collapse
Affiliation(s)
- Jianda Yuan
- Memorial Sloan-Kettering Cancer Center, 1275 New York Ave Box 386, New York, NY 10065 USA
| | - Priti S Hegde
- Genentech, Inc., 1 DNA Way South, San Francisco, CA 94080 USA
| | - Raphael Clynes
- Bristol-Myers Squibb, 3551 Lawrenceville Road, Princeton, NJ 08648 USA
| | - Periklis G Foukas
- Center of Experimental Therapeutics and Ludwig Institute of Cancer Research, University Hospital of Lausanne, Rue du Bugnon 21, 1011 Lausanne, Switzerland ; Department of Pathology, University of Athens Medical School, "Attikon" University Hospital, 1st Rimini St, 12462 Haidari, Greece
| | - Alexandre Harari
- Center of Experimental Therapeutics and Ludwig Institute of Cancer Research, University Hospital of Lausanne, Rue du Bugnon 21, 1011 Lausanne, Switzerland
| | - Thomas O Kleen
- Epiontis GmbH, Rudower Chaussee 29, 12489 Berlin, Germany
| | - Pia Kvistborg
- Netherlands Cancer Institute, Postbus 90203, 1006 BE Amsterdam, Netherlands
| | - Cristina Maccalli
- Italian Network for Biotherapy of Tumors (NIBIT)-Laboratory, c/o Medical Oncology and Immunotherapy, University Hospital of Siena, V.le Bracci,16, Siena, 53100 Italy
| | - Holden T Maecker
- Stanford University Medical Center, 299 Campus Drive, Stanford, CA 94303 USA
| | - David B Page
- Earle A. Chiles Research Institute, Providence Cancer Center, 4805 NE Glisan Street, Portland, OR 97213 USA
| | - Harlan Robins
- Adaptive Technologies, Inc., 1551 Eastlake Avenue East Suite 200, Seattle, WA 98102 USA
| | - Wenru Song
- AstraZeneca, One MedImmune Way, Gaithersburg, MD 20878 USA
| | | | - Ena Wang
- Sidra Medical and Research Center, PO Box 26999, Doha, Qatar
| | - Theresa L Whiteside
- University of Pittsburgh Cancer Institute, 5117 Centre Ave, Suite 1.27, Pittsburgh, PA 15213 USA
| | - Yingdong Zhao
- National Cancer Institute, 9609 Medical Center Drive, Rockville, MD 20850 USA
| | - Heinz Zwierzina
- Innsbruck Medical University, Medizinische Klinik, Anichstrasse 35, Innsbruck, A-6020 Austria
| | - Lisa H Butterfield
- Department of Medicine, Surgery and Immunology, University of Pittsburgh Cancer Institute, 5117 Centre Avenue, Pittsburgh, PA 15213 USA
| | - Bernard A Fox
- Earle A. Chiles Research Institute, Providence Cancer Center, 4805 NE Glisan Street, Portland, OR 97213 USA
| |
Collapse
|
6013
|
Diem S, Kasenda B, Spain L, Martin-Liberal J, Marconcini R, Gore M, Larkin J. Serum lactate dehydrogenase as an early marker for outcome in patients treated with anti-PD-1 therapy in metastatic melanoma. Br J Cancer 2016; 114:256-61. [PMID: 26794281 PMCID: PMC4742588 DOI: 10.1038/bjc.2015.467] [Citation(s) in RCA: 244] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Revised: 11/12/2015] [Accepted: 12/07/2015] [Indexed: 01/20/2023] Open
Abstract
Background: Treatment with programmed death receptor-1 (PD-1) antibodies is associated with high response rates in patients with advanced melanoma. Reliable markers for early response and outcome are still sparse. Methods: We evaluated 66 consecutive patients with advanced/metastatic melanoma treated with nivolumab or pembrolizumab between 2013 and 2014. The main objectives of this study were to investigate whether, first, serum lactate dehydrogenase (LDH) at baseline (normal vs above the upper limit of normal) correlates with overall survival (OS), and, second, whether the change of LDH during treatment predicts response before the first scan and OS in patients with an elevated baseline LDH. Results: After a median follow-up of 9 months, patients with an elevated baseline LDH (N=34) had a significantly shorter OS compared with patients with normal LDH (N=32; 6-month OS: 60.8% vs 81.6% and 12-month OS: 44.2% vs 71.5% (log-rank P=0.0292). In those 34 patients with elevated baseline LDH, the relative change during treatment was significantly associated with an objective response on the first scan: the 11 (32%) patients with partial remission had a mean reduction of −27.3% from elevated baseline LDH. In contrast, patients with progressive disease (N=15) had a mean increase of +39%. Patients with a relative increase over 10% from elevated baseline LDH had a significantly shorter OS compared with patients with ⩽10% change (4.3 vs 15.7 months, log-rank P<0.00623). Conclusions: LDH could be a useful marker at baseline and during treatment to predict early response or progression in patients with advanced melanoma who receive anti-PD-1 therapy.
Collapse
Affiliation(s)
- S Diem
- Department of Medical Oncology, Royal Marsden Hospital NHS Foundation Trust, Fulham Road, London SW36JJ, UK
| | - B Kasenda
- Department of Medical Oncology, Royal Marsden Hospital NHS Foundation Trust, Fulham Road, London SW36JJ, UK
| | - L Spain
- Department of Medical Oncology, Royal Marsden Hospital NHS Foundation Trust, Fulham Road, London SW36JJ, UK
| | - J Martin-Liberal
- Department of Medical Oncology, Royal Marsden Hospital NHS Foundation Trust, Fulham Road, London SW36JJ, UK.,Department of Medical Oncology, Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology (VHIO), Pg Vall d'Hebron, 119-129, 08035 Barcelona, Spain
| | - R Marconcini
- Department of Medical Oncology, Santa Chiara Hospital, via Roma 67, 56100 Pisa, Italy
| | - M Gore
- Department of Medical Oncology, Royal Marsden Hospital NHS Foundation Trust, Fulham Road, London SW36JJ, UK
| | - J Larkin
- Department of Medical Oncology, Royal Marsden Hospital NHS Foundation Trust, Fulham Road, London SW36JJ, UK
| |
Collapse
|
6014
|
|
6015
|
Kourie HR, Awada G, Awada AH. Unknown primary tumors: is there a future therapeutic role for immune checkpoint inhibitors? Future Oncol 2016; 12:429-31. [PMID: 26776661 DOI: 10.2217/fon.15.329] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Hampig Raphael Kourie
- Medical Oncology Clinic, Jules Bordet Institute, Université Libre de Bruxelles, Brussels, Belgium
| | - Gil Awada
- Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Ahmad Hussein Awada
- Medical Oncology Clinic, Jules Bordet Institute, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
6016
|
Yuan J, Hegde PS, Clynes R, Foukas PG, Harari A, Kleen TO, Kvistborg P, Maccalli C, Maecker HT, Page DB, Robins H, Song W, Stack EC, Wang E, Whiteside TL, Zhao Y, Zwierzina H, Butterfield LH, Fox BA. Novel technologies and emerging biomarkers for personalized cancer immunotherapy. J Immunother Cancer 2016; 4:3. [PMID: 26788324 PMCID: PMC4717548 DOI: 10.1186/s40425-016-0107-3] [Citation(s) in RCA: 155] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 01/05/2016] [Indexed: 12/13/2022] Open
Abstract
The culmination of over a century’s work to understand the role of the immune system in tumor control has led to the recent advances in cancer immunotherapies that have resulted in durable clinical responses in patients with a variety of malignancies. Cancer immunotherapies are rapidly changing traditional treatment paradigms and expanding the therapeutic landscape for cancer patients. However, despite the current success of these therapies, not all patients respond to immunotherapy and even those that do often experience toxicities. Thus, there is a growing need to identify predictive and prognostic biomarkers that enhance our understanding of the mechanisms underlying the complex interactions between the immune system and cancer. Therefore, the Society for Immunotherapy of Cancer (SITC) reconvened an Immune Biomarkers Task Force to review state of the art technologies, identify current hurdlers, and make recommendations for the field. As a product of this task force, Working Group 2 (WG2), consisting of international experts from academia and industry, assembled to identify and discuss promising technologies for biomarker discovery and validation. Thus, this WG2 consensus paper will focus on the current status of emerging biomarkers for immune checkpoint blockade therapy and discuss novel technologies as well as high dimensional data analysis platforms that will be pivotal for future biomarker research. In addition, this paper will include a brief overview of the current challenges with recommendations for future biomarker discovery.
Collapse
Affiliation(s)
- Jianda Yuan
- Memorial Sloan-Kettering Cancer Center, 1275 New York Ave Box 386, New York, NY 10065 USA
| | - Priti S Hegde
- Genentech, Inc., 1 DNA Way South, San Francisco, CA 94080 USA
| | - Raphael Clynes
- Bristol-Myers Squibb, 3551 Lawrenceville Road, Princeton, NJ 08648 USA
| | - Periklis G Foukas
- Center of Experimental Therapeutics and Ludwig Institute of Cancer Research, University Hospital of Lausanne, Rue du Bugnon 21, 1011 Lausanne, Switzerland ; Department of Pathology, University of Athens Medical School, "Attikon" University Hospital, 1st Rimini St, 12462 Haidari, Greece
| | - Alexandre Harari
- Center of Experimental Therapeutics and Ludwig Institute of Cancer Research, University Hospital of Lausanne, Rue du Bugnon 21, 1011 Lausanne, Switzerland
| | - Thomas O Kleen
- Epiontis GmbH, Rudower Chaussee 29, 12489 Berlin, Germany
| | - Pia Kvistborg
- Netherlands Cancer Institute, Postbus 90203, 1006 BE Amsterdam, Netherlands
| | - Cristina Maccalli
- Italian Network for Biotherapy of Tumors (NIBIT)-Laboratory, c/o Medical Oncology and Immunotherapy, University Hospital of Siena, V.le Bracci,16, Siena, 53100 Italy
| | - Holden T Maecker
- Stanford University Medical Center, 299 Campus Drive, Stanford, CA 94303 USA
| | - David B Page
- Earle A. Chiles Research Institute, Providence Cancer Center, 4805 NE Glisan Street, Portland, OR 97213 USA
| | - Harlan Robins
- Adaptive Technologies, Inc., 1551 Eastlake Avenue East Suite 200, Seattle, WA 98102 USA
| | - Wenru Song
- AstraZeneca, One MedImmune Way, Gaithersburg, MD 20878 USA
| | | | - Ena Wang
- Sidra Medical and Research Center, PO Box 26999, Doha, Qatar
| | - Theresa L Whiteside
- University of Pittsburgh Cancer Institute, 5117 Centre Ave, Suite 1.27, Pittsburgh, PA 15213 USA
| | - Yingdong Zhao
- National Cancer Institute, 9609 Medical Center Drive, Rockville, MD 20850 USA
| | - Heinz Zwierzina
- Innsbruck Medical University, Medizinische Klinik, Anichstrasse 35, Innsbruck, A-6020 Austria
| | - Lisa H Butterfield
- Department of Medicine, Surgery and Immunology, University of Pittsburgh Cancer Institute, 5117 Centre Avenue, Pittsburgh, PA 15213 USA
| | - Bernard A Fox
- Earle A. Chiles Research Institute, Providence Cancer Center, 4805 NE Glisan Street, Portland, OR 97213 USA
| |
Collapse
|
6017
|
Lester-Coll NH, Rutter CE, Bledsoe TJ, Goldberg SB, Decker RH, Yu JB. Cost-Effectiveness of Surgery, Stereotactic Body Radiation Therapy, and Systemic Therapy for Pulmonary Oligometastases. Int J Radiat Oncol Biol Phys 2016; 95:663-72. [PMID: 27055395 DOI: 10.1016/j.ijrobp.2016.01.020] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 01/06/2016] [Accepted: 01/12/2016] [Indexed: 02/03/2023]
Abstract
INTRODUCTION Pulmonary oligometastases have conventionally been managed with surgery and/or systemic therapy. However, given concerns about the high cost of systemic therapy and improvements in local treatment of metastatic cancer, the optimal cost-effective management of these patients is unclear. Therefore, we sought to assess the cost-effectiveness of initial management strategies for pulmonary oligometastases. METHODS AND MATERIALS A cost-effectiveness analysis using a Markov modeling approach was used to compare average cumulative costs, quality adjusted life years (QALYs), and incremental cost-effectiveness ratios (ICERs) among 3 initial disease management strategies: video-assisted thoracic surgery (VATS) wedge resection, stereotactic body radiation therapy (SBRT), and systemic therapy among 5 different cohorts of patient disease: (1) melanoma; (2) non-small cell lung cancer adenocarcinoma without an EGFR mutation (NSCLC AC); (3) NSCLC with an EGFR mutation (NSCLC EGFRm AC); (4) NSCLC squamous cell carcinoma (NSCLC SCC); and (5) colon cancer. One-way sensitivity analyses and probabilistic sensitivity analyses were performed to analyze uncertainty with regard to model parameters. RESULTS In the base case, SBRT was cost effective for melanoma, with costs/net QALYs of $467,787/0.85. In patients with NSCLC, the most cost-effective strategies were SBRT for AC ($156,725/0.80), paclitaxel/carboplatin for SCC ($123,799/0.48), and erlotinib for EGFRm AC ($147,091/1.90). Stereotactic body radiation therapy was marginally cost-effective for EGFRm AC compared to erlotinib with an incremental cost-effectiveness ratio of $126,303/QALY. For colon cancer, VATS wedge resection ($147,730/2.14) was the most cost-effective strategy. Variables with the greatest influence in the model were erlotinib-associated progression-free survival (EGFRm AC), toxicity (EGFRm AC), cost of SBRT (NSCLC SCC), and patient utilities (all histologies). CONCLUSIONS Video-assisted thoracic surgery wedge resection or SBRT can be cost-effective in select patients with pulmonary oligometastases, depending on histology, efficacy, and tolerability of treatment and patient preferences.
Collapse
Affiliation(s)
- Nataniel H Lester-Coll
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, Connecticut.
| | - Charles E Rutter
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, Connecticut
| | - Trevor J Bledsoe
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, Connecticut
| | - Sarah B Goldberg
- Department of Medicine (Medical Oncology), Yale University School of Medicine, New Haven, Connecticut
| | - Roy H Decker
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, Connecticut
| | - James B Yu
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
6018
|
Matter A. Bridging academic science and clinical research in the search for novel targeted anti-cancer agents. Cancer Biol Med 2016; 12:316-27. [PMID: 26779369 PMCID: PMC4706519 DOI: 10.7497/j.issn.2095-3941.2015.0079] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
This review starts with a brief history of drug discovery & development, and the place of Asia in this worldwide effort discussed. The conditions and constraints of a successful translational R&D involving academic basic research and clinical research are discussed and the Singapore model for pursuit of open R&D described. The importance of well-characterized, validated drug targets for the search for novel targeted anti-cancer agents is emphasized, as well as a structured, high quality translational R&D. Furthermore, the characteristics of an attractive preclinical development drug candidate are discussed laying the foundation of a successful preclinical development. The most frequent sources of failures are described and risk management at every stage is highly recommended. Organizational factors are also considered to play an important role. The factors to consider before starting a new drug discovery & development project are described, and an example is given of a successful clinical project that has had its roots in local universities and was carried through preclinical development into phase I clinical trials.
Collapse
Affiliation(s)
- Alex Matter
- Experimental Therapeutics Center & D3, ASTAR, Singapore 138669, Singapore
| |
Collapse
|
6019
|
Abdel-Rahman O, ElHalawani H, Fouad M. Risk of endocrine complications in cancer patients treated with immune check point inhibitors: a meta-analysis. Future Oncol 2016; 12:413-25. [PMID: 26775673 DOI: 10.2217/fon.15.222] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND We performed a meta-analysis of the risk of endocrine adverse events associated with immune check point inhibitors. METHODS Eligible studies included randomized trials of cancer patients on immune checkpoint inhibitors; describing events of hypothyroidism, hyperthyroidism, hypophysitis and adrenal insufficiency. RESULTS A total of ten clinical trials were eligible for the meta-analysis. The relative risk of all-grade hypothyroidism, hyperthyroidism, hypophyisitis and adrenal insufficiency were 8.26 (95% CI: 4.67-14.62; p < 0.00001), 5.48 (95% CI: 1.33-22.53; p = 0.02); 22.03 (95% CI: 8.52-56.94; p < 0.00001), 3.87 (95% CI: 1.12-13.41; p = 0.03), respectively. CONCLUSION Our meta-analysis has demonstrated that the use of immune check point inhibitors is associated with an increased risk of hypothyroidism, hyperthyroidism, hypophysitis and adrenal insufficiency compared with control.
Collapse
Affiliation(s)
- Omar Abdel-Rahman
- Clinical Oncology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Hesham ElHalawani
- Clinical Oncology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Mona Fouad
- Medical Microbiology & Immunology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| |
Collapse
|
6020
|
Potential biomarker for checkpoint blockade immunotherapy and treatment strategy. Tumour Biol 2016; 37:4251-61. [PMID: 26779629 DOI: 10.1007/s13277-016-4812-9] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 01/06/2016] [Indexed: 12/12/2022] Open
Abstract
Programmed cell death protein-1 (PD-1) and ligand (PD-L1) provide an important escape mechanism from immune attack, and blockade therapy of these proteins show promising clinical benefits in many types of cancer. PD-L1 can be induced by interferon-gamma (IFN-γ), hypoxia, or toll-like receptor (TLR)-mediated pathways that confer adaptive immune resistance, or upregulated by oncogenic signals leading to constitutive expression and resulting in intrinsic immune resistance. The PD-1/PD-L1 checkpoint blockade, which targets regulatory pathways in T cells to overcome immune resistance, is correlated to PD-L1 expression pattern and the presence of tumor-infiltrating lymphocytes (TILs). Meanwhile, immunogenic mutation loads show significant response to checkpoint blockade, which is probably due to PD-1/L1 status and TIL content. Finally, the clinical strategies to design effective checkpoint-targeting immunotherapies are based on the classification of inducible/constitutive expression of PD-L1 and the presence of TILs.
Collapse
|
6021
|
Tumour-infiltrating lymphocytes in melanoma prognosis and cancer immunotherapy. Pathology 2016; 48:177-87. [PMID: 27020390 DOI: 10.1016/j.pathol.2015.12.006] [Citation(s) in RCA: 194] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2015] [Accepted: 11/16/2015] [Indexed: 12/26/2022]
Abstract
The field of systemic cancer therapy for metastatic disease has entered an exciting era with the advent of novel immunomodulatory strategies targeting immune checkpoints. At the heart of these promising efforts are the tumour-infiltrating lymphocytes (TILs). As the reports demonstrating efficacy of modulating TIL effector function in patients with advanced stage cancer continue to accrue, it has become essential to better understand TIL immunobiology in order to further improve clinical outcome. In addition to providing an overview of the current immunotherapies available for metastatic melanoma, this review will briefly introduce the history and classification of TILs. Moreover, we will dissect the multifaceted roles of TILs in tumour-specific immunity and melanoma immune escape. The significance of TILs in melanoma prognosis and cancer immunotherapy will also be discussed, with a particular focus on their potential utility as biomarkers of patient response. The goal of personalised medicine appears to be in realistic sight, as new immunomodulatory techniques and technological innovations continue to advance the field of cancer immunotherapy. In light of recent studies highlighting the possible utility of TILs in determining therapeutic outcome, further characterisation of TIL phenotype and function has the potential to help translate individualised care into current medical practice.
Collapse
|
6022
|
Singh BP, Salama AKS. Updates in Therapy for Advanced Melanoma. Cancers (Basel) 2016; 8:E17. [PMID: 26784231 PMCID: PMC4728464 DOI: 10.3390/cancers8010017] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Revised: 01/04/2016] [Accepted: 01/07/2016] [Indexed: 01/15/2023] Open
Abstract
Cutaneous melanoma is one of the most aggressive forms of skin cancer, and is correlated with a large proportion of skin cancer-related deaths. Therapy for cutaneous melanoma has advanced greatly through careful identification of therapeutic targets and the development of novel immunotherapeutic approaches. The identification of BRAF as well as other driver mutations, have allowed for a specialized approach to treatment. In addition, immune checkpoint inhibition has dramatically changed the treatment landscape over the past 5-10 years. The successful targeting of CTLA-4, as well as PD-1/PD-L1, has been translated into meaningful clinical benefit for patients, with multiple other potential agents in development. Systemic therapy for cutaneous melanoma is becoming more nuanced and often takes a multifaceted strategy. This review aims to discuss the benefits and limitations of current therapies in systemic melanoma treatment as well as areas of future development.
Collapse
Affiliation(s)
- Bhavana P Singh
- Department of Internal Medicine, Duke University Medical Center, Durham, NC 27710, USA.
| | - April K S Salama
- Division of Medical Oncology, Department of Internal Medicine, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
6023
|
Abstract
As new active agents are discovered and approved, it is clear that combinations of these immunotherapies will be needed to reject most human cancers. The era of personalized patient tumor analysis has arrived just in time to guide these combinations.
Collapse
Affiliation(s)
- James C Yang
- Surgery Branch, National Cancer Institute, Bethesda, Maryland.
| |
Collapse
|
6024
|
Chatzistefanou I, Kolokythas A, Vahtsevanos K, Antoniades K. Primary mucosal melanoma of the oral cavity: current therapy and future directions. Oral Surg Oral Med Oral Pathol Oral Radiol 2016; 122:17-27. [PMID: 27039005 DOI: 10.1016/j.oooo.2016.01.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Revised: 01/04/2016] [Accepted: 01/05/2016] [Indexed: 02/03/2023]
Abstract
BACKGROUND Primary mucosal melanoma of the oral cavity is a highly aggressive malignancy of melanocytic origin. The aim of this study is to report a case series of oral mucosal melanomas (OMMs) and provide a review of the literature with regard to treatment guidelines for and prognosis of this pathologic entity. STUDY DESIGN We report three cases of OMMs treated in our institutions and the results of a literature review, in which the words "oral" and "mucosal melanoma" were used as the main keywords. RESULTS Surgical resection of the primary tumor with wide resection margins appears to be the recommended primary treatment modality with the aim to achieve tumor-free margins. Elective neck dissection and adjuvant radiotherapy have been advocated for locoregional control. Chemotherapy has not been shown to improve survival and is mainly used for palliative purposes. Immunotherapy and biochemotherapy seem to significantly improve survival and could open new therapeutic horizons. CONCLUSIONS The prognosis of OMMs remains poor despite treatment. Early diagnosis and aggressive surgical resection followed by adjuvant therapies could be the key to improving survival. Multicenter randomized clinical trials, which may be difficult to conduct because of the rarity to the lesion, would aid in the development of new strategies.
Collapse
Affiliation(s)
- Ioannis Chatzistefanou
- Department of Oral and Maxillofacial Surgery, Aristotle University of Thessaloniki, "G. Papanikolaou" General Hospital, Thessaloniki, Greece
| | - Antonia Kolokythas
- Department of Oral and Maxillofacial Surgery, University of Rochester, Rochester, New York, USA.
| | - Konstantinos Vahtsevanos
- Department of Oral and Maxillofacial Surgery, Aristotle University of Thessaloniki, "G. Papanikolaou" General Hospital, Thessaloniki, Greece
| | - Konstantinos Antoniades
- Department of Oral and Maxillofacial Surgery, University of Rochester, Rochester, New York, USA
| |
Collapse
|
6025
|
Kong Y, Zhu L, Schell TD, Zhang J, Claxton DF, Ehmann WC, Rybka WB, George MR, Zeng H, Zheng H. T-Cell Immunoglobulin and ITIM Domain (TIGIT) Associates with CD8+ T-Cell Exhaustion and Poor Clinical Outcome in AML Patients. Clin Cancer Res 2016; 22:3057-66. [PMID: 26763253 DOI: 10.1158/1078-0432.ccr-15-2626] [Citation(s) in RCA: 231] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 01/04/2016] [Indexed: 11/16/2022]
Abstract
PURPOSE T-cell immunoglobulin and immunoreceptor tyrosine-based inhibitory motif (ITIM) domain (TIGIT) is a recently identified T-cell coinhibitory receptor. In this study, we aimed to determine the clinical impact of TIGIT in patients with acute myelogenous leukemia (AML) and dissect the role of TIGIT in the pathogenesis of leukemia progression. EXPERIMENTAL DESIGN TIGIT expression on T cells from peripheral blood collected from patients with AML was examined by flow cytometry. The correlation of TIGIT expression to clinical outcomes, including rate of complete remission and relapse post-allogeneic stem cell transplantation (alloSCT) in AML patients, was analyzed. Phenotypic and functional study (cytokine release, proliferation, killing, and apoptosis) of TIGIT-expressing T cells were performed. Using siRNA to silence TIGIT, we further elucidated the regulatory role of TIGIT in the T-cell immune response by dissecting the effect of TIGIT knockdown on cytokine release and apoptosis of T cells from AML patients. RESULTS TIGIT expression on CD8(+) T cells is elevated in AML patients and high-TIGIT correlates with primary refractory disease and leukemia relapse post-alloSCT. TIGIT(+) CD8(+) T cells display phenotypic features of exhaustion and exhibit functional impairment manifested by low production of cytokines and high susceptibility to apoptosis. Importantly, their functional defects are reversed by TIGIT knockdown. CONCLUSIONS TIGIT contributes to functional T-cell impairment and associates with poor clinical outcome in AML. Our study suggests that blockade of TIGIT to restore T-cell function and antitumor immunity may represent a novel effective leukemia therapeutic. Clin Cancer Res; 22(12); 3057-66. ©2016 AACR.
Collapse
Affiliation(s)
- Yaxian Kong
- Penn State Hershey Cancer Institute, Penn State University College of Medicine, Hershey, Pennsylvania. Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing Key Laboratory of Emerging Infectious Diseases, Beijing, China
| | - Liuluan Zhu
- Penn State Hershey Cancer Institute, Penn State University College of Medicine, Hershey, Pennsylvania. Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing Key Laboratory of Emerging Infectious Diseases, Beijing, China
| | - Todd D Schell
- Penn State Hershey Cancer Institute, Penn State University College of Medicine, Hershey, Pennsylvania. Department of Microbiology and Immunology, Penn State University College of Medicine, Hershey, Pennsylvania
| | - Jianhong Zhang
- Penn State Hershey Cancer Institute, Penn State University College of Medicine, Hershey, Pennsylvania
| | - David F Claxton
- Penn State Hershey Cancer Institute, Penn State University College of Medicine, Hershey, Pennsylvania
| | - W Christopher Ehmann
- Penn State Hershey Cancer Institute, Penn State University College of Medicine, Hershey, Pennsylvania
| | - Witold B Rybka
- Penn State Hershey Cancer Institute, Penn State University College of Medicine, Hershey, Pennsylvania
| | - Melissa R George
- Department of Pathology, Penn State Hershey Medical Center, Penn State University College of Medicine, Hershey, Pennsylvania
| | - Hui Zeng
- Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing Key Laboratory of Emerging Infectious Diseases, Beijing, China.
| | - Hong Zheng
- Penn State Hershey Cancer Institute, Penn State University College of Medicine, Hershey, Pennsylvania.
| |
Collapse
|
6026
|
Innovative perspectives of immunotherapy in head and neck cancer. From relevant scientific rationale to effective clinical practice. Cancer Treat Rev 2016; 43:113-23. [PMID: 26827699 DOI: 10.1016/j.ctrv.2016.01.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Revised: 12/17/2015] [Accepted: 01/06/2016] [Indexed: 01/05/2023]
Abstract
It is now well established that head and neck cancer carcinogenesis is characterized by genetic instability and several immune defects, leading to unique host-tumor interactions. In such condition, recent improved comprehension and relevant findings could lead to identification of innovative molecular therapeutic targets, achieving considerable clinical and translational research. This review aims to summarize and to highlight most recent and relevant scientific rationale in this era of immunotherapy revival, and to correlate it to the near future clinical practice for the management of this challenging disease.
Collapse
|
6027
|
Ajithkumar T, Parkinson C, Fife K, Corrie P, Jefferies S. Evolving treatment options for melanoma brain metastases. Lancet Oncol 2016; 16:e486-97. [PMID: 26433822 DOI: 10.1016/s1470-2045(15)00141-2] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2015] [Revised: 07/04/2015] [Accepted: 07/08/2015] [Indexed: 11/28/2022]
Abstract
Melanoma is a leading cause of lost productivity due to premature cancer mortality. Melanoma frequently spreads to the brain and is associated with rapid deterioration in quality and quantity of life. Until now, treatment options have been restricted to surgery and radiotherapy, although neither modality has been well studied in clinical trials. However, the new immune checkpoint inhibitors and molecularly targeted agents that have been introduced for treatment of metastatic melanoma are active against brain metastases and offer new opportunities to improve disease outcomes. New challenges arise, including how to integrate or sequence multiple treatment modalities, and current practice varies widely. In this Review, we summarise evidence for the treatment of melanoma brain metastases, and discuss the rationale and evidence for combination modalities, highlighting areas for future research.
Collapse
Affiliation(s)
- Thankamma Ajithkumar
- Department of Oncology, Cambridge University Hospitals National Health Service Foundation Trust, Cambridge, UK.
| | - Christine Parkinson
- Department of Oncology, Cambridge University Hospitals National Health Service Foundation Trust, Cambridge, UK
| | - Kate Fife
- Department of Oncology, Cambridge University Hospitals National Health Service Foundation Trust, Cambridge, UK
| | - Pippa Corrie
- Department of Oncology, Cambridge University Hospitals National Health Service Foundation Trust, Cambridge, UK
| | - Sarah Jefferies
- Department of Oncology, Cambridge University Hospitals National Health Service Foundation Trust, Cambridge, UK
| |
Collapse
|
6028
|
Hwang SJE, Carlos G, Wakade D, Byth K, Kong BY, Chou S, Carlino MS, Kefford R, Fernandez-Penas P. Cutaneous adverse events (AEs) of anti-programmed cell death (PD)-1 therapy in patients with metastatic melanoma: A single-institution cohort. J Am Acad Dermatol 2016; 74:455-61.e1. [PMID: 26793994 DOI: 10.1016/j.jaad.2015.10.029] [Citation(s) in RCA: 228] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Revised: 10/13/2015] [Accepted: 10/15/2015] [Indexed: 12/11/2022]
Abstract
BACKGROUND Anti-programmed cell death (PD)-1 therapy is emerging as the backbone of new standard of care immunotherapy for metastatic melanoma. Immune-related cutaneous events are observed in these patients. OBJECTIVE We sought to describe cutaneous adverse events observed in patients with metastatic melanoma on anti-PD-1 therapy. METHODS We reviewed the clinical and histologic information of all patients treated with single-agent anti-PD-1 therapy for metastatic melanoma at Westmead Hospital, Sydney, Australia, from May 2012 to February 2015. RESULTS Of the 82 patients included in the study, 34 had dermatology assessments. Forty (49%) developed a form of anti-PD-1-associated cutaneous adverse events. In all, 17% developed lichenoid reactions and eczema, and 15% developed vitiligo. An estimated 25% of patients were expected to develop their first lichenoid reactions within 8.3 months, and eczema and vitiligo within 10.3 months of therapy. These adverse events tend to appear together in patients on anti-PD-1 therapy. LIMITATIONS The study was from a single center and clinical information was reviewed retrospectively in patients not referred to dermatology. CONCLUSION Anti-PD-1 therapy is associated with the development of immune-related cutaneous events. Lichenoid reactions, eczema, and vitiligo are the 3 most prevalent lesions observed in our population. There is a tendency for lichenoid reactions and eczema to occur with vitiligo.
Collapse
Affiliation(s)
- Shelley Ji Eun Hwang
- Department of Dermatology, Westmead Hospital, Sydney, Australia; Sydney Medical School, University of Sydney, Sydney, Australia.
| | - Giuliana Carlos
- Department of Dermatology, Westmead Hospital, Sydney, Australia; Sydney Medical School, University of Sydney, Sydney, Australia
| | - Deepal Wakade
- Department of Dermatology, Westmead Hospital, Sydney, Australia; Sydney Medical School, University of Sydney, Sydney, Australia
| | - Karen Byth
- Research and Education Network, Westmead Hospital, Sydney, Australia; Sydney Medical School, University of Sydney, Sydney, Australia
| | - Benjamin Y Kong
- Crown Princess Mary Cancer Center, Westmead Hospital, Sydney, Australia
| | - Shaun Chou
- Tissue Pathology and Diagnostic Oncology, Westmead Hospital, Sydney, Australia
| | - Matteo S Carlino
- Crown Princess Mary Cancer Center, Westmead Hospital, Sydney, Australia; Sydney Medical School, University of Sydney, Sydney, Australia; Melanoma Institute Australia, Sydney, Australia
| | - Richard Kefford
- Crown Princess Mary Cancer Center, Westmead Hospital, Sydney, Australia; Melanoma Institute Australia, Sydney, Australia
| | - Pablo Fernandez-Penas
- Department of Dermatology, Westmead Hospital, Sydney, Australia; Sydney Medical School, University of Sydney, Sydney, Australia
| |
Collapse
|
6029
|
Sano T, Uhara H, Mikoshiba Y, Kobayashi A, Uchiyama R, Tateishi K, Yamamoto H, Okuyama R. Nivolumab-induced organizing pneumonia in a melanoma patient. Jpn J Clin Oncol 2016; 46:270-2. [PMID: 26759348 DOI: 10.1093/jjco/hyv199] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 12/05/2015] [Indexed: 11/14/2022] Open
Abstract
We report the case of a 70-year-old woman with vaginal melanoma and multiple metastases in the lung. After the third dose of nivolumab, decreased room-air resting arterial oxygen saturation with bilateral basal fine crackles on auscultation developed despite the absence of respiratory symptoms. Computed tomography showed ground-glass opacities with airspace consolidations scattered with a peculiar distribution, and most were observed around the existing metastatic tumors in the lung. From the 42nd day to the 56th day after the last administration of nivolumab, she received dexamethasone 1-2 mg/body for the prevention of adverse events after stereotactic radiation for brain metastasis. At 3 months after the last administration of nivolumab, a computed tomography scan revealed improvement of the pneumonia and a decreased size and number of metastatic lesions in the lung, although some lesions showed enlargement. Further examination is needed to clarify the relationship between the pattern of pneumonia after Nivo therapy and clinical effects.
Collapse
Affiliation(s)
- Tasuku Sano
- Department of Dermatology, Shinshu University School of Medicine, Matsumoto
| | - Hisashi Uhara
- Department of Dermatology, Shinshu University School of Medicine, Matsumoto
| | - Yasutomo Mikoshiba
- Department of Dermatology, Shinshu University School of Medicine, Matsumoto
| | - Aya Kobayashi
- Department of Dermatology, Shinshu University School of Medicine, Matsumoto
| | - Ryuhei Uchiyama
- Department of Dermatology, Shinshu University School of Medicine, Matsumoto
| | - Kazunari Tateishi
- First Department of Internal Medicine, Shinshu University School of Medicine, Matsumoto, Japan
| | - Hiroshi Yamamoto
- First Department of Internal Medicine, Shinshu University School of Medicine, Matsumoto, Japan
| | - Ryuhei Okuyama
- Department of Dermatology, Shinshu University School of Medicine, Matsumoto
| |
Collapse
|
6030
|
Wurz GT, Kao CJ, DeGregorio MW. Novel cancer antigens for personalized immunotherapies: latest evidence and clinical potential. Ther Adv Med Oncol 2016; 8:4-31. [PMID: 26753003 DOI: 10.1177/1758834015615514] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The clinical success of monoclonal antibody immune checkpoint modulators such as ipilimumab, which targets cytotoxic T lymphocyte-associated antigen 4 (CTLA-4), and the recently approved agents nivolumab and pembrolizumab, which target programmed cell death receptor 1 (PD-1), has stimulated renewed enthusiasm for anticancer immunotherapy, which was heralded by Science as 'Breakthrough of the Year' in 2013. As the potential of cancer immunotherapy has been recognized since the 1890s when William Coley showed that bacterial products could be beneficial in cancer patients, leveraging the immune system in the treatment of cancer is certainly not a new concept; however, earlier attempts to develop effective therapeutic vaccines and antibodies against solid tumors, for example, melanoma, frequently met with failure due in part to self-tolerance and the development of an immunosuppressive tumor microenvironment. Increased knowledge of the mechanisms through which cancer evades the immune system and the identification of tumor-associated antigens (TAAs) and negative immune checkpoint regulators have led to the development of vaccines and monoclonal antibodies targeting specific tumor antigens and immune checkpoints such as CTLA-4 and PD-1. This review first discusses the established targets of currently approved cancer immunotherapies and then focuses on investigational cancer antigens and their clinical potential. Because of the highly heterogeneous nature of tumors, effective anticancer immunotherapy-based treatment regimens will likely require a personalized combination of therapeutic vaccines, antibodies and chemotherapy that fit the specific biology of a patient's disease.
Collapse
Affiliation(s)
- Gregory T Wurz
- Department of Internal Medicine, Division of Hematology and Oncology, University of California, Davis, Sacramento, CA, USA
| | - Chiao-Jung Kao
- Department of Obstetrics and Gynecology, University of California, Davis Sacramento, CA, USA
| | - Michael W DeGregorio
- Department of Internal Medicine, Division of Hematology and Oncology, University of California, Davis, 4501 X Street Suite 3016, Sacramento, CA 95817, USA
| |
Collapse
|
6031
|
Abstract
T cell checkpoint blockade therapies are revolutionizing the treatment of patients with cancer. Highlighted by the recent success of PD-1 plus CTLA-4 blockade in patients with melanomas, synergistic immunotherapy combinations of modalities represent an important opportunity to improve responses and outcomes for patients. We review the rationale and experience with T cell checkpoint blockade in combination with targeting of other coinhibitory or costimulatory checkpoints, immunomodulatory molecules in the tumor microenvironment, and other anticancer modalities such as vaccines, chemotherapy, and radiation.
Collapse
Affiliation(s)
- Matthew D Hellmann
- Memorial Sloan Kettering Cancer Center, New York, NY, United States; Weill Cornell Medical College, New York, NY, United States
| | - Claire F Friedman
- Memorial Sloan Kettering Cancer Center, New York, NY, United States; Weill Cornell Medical College, New York, NY, United States
| | - Jedd D Wolchok
- Memorial Sloan Kettering Cancer Center, New York, NY, United States; Weill Cornell Medical College, New York, NY, United States.
| |
Collapse
|
6032
|
Pantziarka P, Sukhatme V, Bouche G, Meheus L, Sukhatme VP. Repurposing Drugs in Oncology (ReDO)-diclofenac as an anti-cancer agent. Ecancermedicalscience 2016; 10:610. [PMID: 26823679 PMCID: PMC4720497 DOI: 10.3332/ecancer.2016.610] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Indexed: 12/16/2022] Open
Abstract
Diclofenac (DCF) is a well-known and widely used non-steroidal anti-inflammatory drug (NSAID), with a range of actions which are of interest in an oncological context. While there has long been an interest in the use of NSAIDs in chemoprevention, there is now emerging evidence that such drugs may have activity in a treatment setting. DCF, which is a potent inhibitor of COX-2 and prostaglandin E2 synthesis, displays a range of effects on the immune system, the angiogenic cascade, chemo- and radio-sensitivity and tumour metabolism. Both pre-clinical and clinical evidence of these effects, in multiple cancer types, is assessed and summarised and relevant mechanisms of action outlined. Based on this evidence the case is made for further clinical investigation of the anticancer effects of DCF, particularly in combination with other agents - with a range of possible multi-drug and multi-modality combinations outlined in the supplementary materials accompanying the main paper.
Collapse
Affiliation(s)
- Pan Pantziarka
- Anticancer Fund, Brussels, 1853 Strombeek-Bever, Belgium
- The George Pantziarka TP53 Trust, London, UK
| | | | | | - Lydie Meheus
- Anticancer Fund, Brussels, 1853 Strombeek-Bever, Belgium
| | - Vikas P Sukhatme
- GlobalCures, Inc; Newton MA 02459, USA
- Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
6033
|
Abstract
Immunotherapy with immune checkpoint inhibition has been improving the outcomes of patients with many different types of malignancies. Immune checkpoint inhibition has been most extensively studied in patients with advanced melanoma and there are three FDA approved antibodies already widely used in clinical practice (ipilimumab, nivolumab, and pembrolizumab). In this chapter, we review the mechanistic basis behind the development of immune checkpoint blocking antibodies. We then discuss specifics regarding each agent, unique clinical considerations in treating patients with this approach, and future directions, including combination strategies. This chapter is focused on melanoma, but the principles related to this immunotherapy approach are applicable to patients with many types of malignancies.
Collapse
|
6034
|
Abstract
Background: Increasing evidence supporting the role of immune checkpoint blockade in cancer management has been bolstered by recent reports demonstrating significant and durable clinical responses across multiple tumour types, including metastatic urothelial carcinoma (mUC). The majority of these results are achieved via blockade of the programmed death (PD) axis, which like CTLA-4 blockade permits T-cell activation and immune-mediated anti-tumour activity- essentially harnessing the patient’s own immune system to mount an anti-neoplastic response. However, while clinical responses can be striking, our understanding of the biology of immune checkpoint blockade is only beginning to shed light on how to maximize and even improve patient outcomes with immune checkpoint blockade, especially in UC. Methods: We performed a literature review for immune checkpoint blockade with a focus on rationale for checkpoint therapy and outcomes in UC. We also highlight the advances made in other tumour types, with a focus on the recent 2015 meeting of the American Society for Clinical Oncology. Results: In heavily pre-treated UC, trials are suggesting objective response rates above 30% . These impressive results are seen across multiple different tumour types, especially those with high burden of DNA level mutations. Identification of prognostic biomarkers is currently under investigation, in order to improve patient selection. Interestingly, response to PD-1 directed therapy is seen even in patients with no evidence of PD-1 positivity on immunohistochemistry. This has led to the development of enhanced biomarkers including assessing DNA mutation rates and immune gene signatures, to improve patient selection. Conclusions: Immune checkpoint blockade is an exciting cancer treatment modality which is demonstrating impressive clinical results across multiple tumour types. For UC, anti-PD directed therapy represents a much needed treatment in the metastatic, post chemotherapy context. Potential for these agents to have clinical utility in non-metastatic UC is still to be assessed.
Collapse
Affiliation(s)
- S Bidnur
- Vancouver Prostate Centre, University of British Columbia , Vancouver, B.C, Canada
| | - R Savdie
- Vancouver Prostate Centre, University of British Columbia , Vancouver, B.C, Canada
| | - P C Black
- Vancouver Prostate Centre, University of British Columbia , Vancouver, B.C, Canada
| |
Collapse
|
6035
|
Kasenda B, Kühnl A, Chau I. Beginning of a novel frontier: T-cell-directed immune manipulation in lymphomas. Expert Rev Hematol 2016; 9:123-35. [DOI: 10.1586/17474086.2016.1122513] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
6036
|
Lau PKH, Ascierto PA, McArthur G. Melanoma: the intersection of molecular targeted therapy and immune checkpoint inhibition. Curr Opin Immunol 2016; 39:30-8. [PMID: 26765776 DOI: 10.1016/j.coi.2015.12.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Revised: 12/16/2015] [Accepted: 12/16/2015] [Indexed: 10/22/2022]
Abstract
Melanoma is at the forefront of development of systemic therapeutics with both molecular targeted therapies and immune checkpoint inhibitors as cornerstones of treatment. Although responses to molecularly targeted therapy is largely from blockade of oncogenic pathways, evidence is emerging of the immunomodulatory effects from BRAF inhibition. Additionally programmed-death-1 (PD-1) inhibitors have revolutionized the treatment of melanoma and are set to pave future improvements in other solid tumors. Combinations of PD-1 inhibitors with novel immune checkpoints or with molecularly targeted therapies are under investigation and may improve on the considerable progress made.
Collapse
Affiliation(s)
- Peter Kar Han Lau
- Department of Cancer Medicine, Peter MacCallum Cancer Centre, St Andrews Place, East Melbourne, Victoria 3002, Australia
| | - Paolo A Ascierto
- Melanoma, Cancer Immunotherapy, and Innovative Therapy Unit, Istituto Nazionale Tumori Fondazione "G. Pascale", Via Mariano Semmola, 80131 Napoli, Italy.
| | - Grant McArthur
- Department of Cancer Medicine, Peter MacCallum Cancer Centre, St Andrews Place, East Melbourne, Victoria 3002, Australia
| |
Collapse
|
6037
|
Gershenwald JE, Guy GP. Stemming the Rising Incidence of Melanoma: Calling Prevention to Action. J Natl Cancer Inst 2016; 108:djv381. [PMID: 26563358 PMCID: PMC6048594 DOI: 10.1093/jnci/djv381] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2015] [Indexed: 12/16/2022] Open
Affiliation(s)
- Jeffrey E Gershenwald
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Gery P Guy
- Division of Cancer Prevention and Control, Centers for Disease Control and Prevention, Atlanta, Georgia
| |
Collapse
|
6038
|
Yang F, Jin H, Wang J, Sun Q, Yan C, Wei F, Ren X. Adoptive Cellular Therapy (ACT) for Cancer Treatment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 909:169-239. [PMID: 27240459 DOI: 10.1007/978-94-017-7555-7_4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Adoptive cellular therapy (ACT) with various lymphocytes or antigen-presenting cells is one stone in the pillar of cancer immunotherapy, which relies on the tumor-specific T cell. The transfusion of bulk T-cell population into patients is an effective treatment for regression of cancer. In this chapter, we summarize the development of various strategies in ACT for cancer immunotherapy and discuss some of the latest progress and obstacles in technical, safety, and even regulatory aspects to translate these technologies to the clinic. ACT is becoming a potentially powerful approach to cancer treatment. Further experiments and clinical trials are needed to optimize this strategy.
Collapse
Affiliation(s)
- Fan Yang
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, Tianjin, China
- Department of Biotherapy, Tianjin Medical University Cancer Institute and Hospital, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, Tianjin, China
| | - Hao Jin
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, Tianjin, China
| | - Jian Wang
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, Tianjin, China
| | - Qian Sun
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, Tianjin, China
| | - Cihui Yan
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, Tianjin, China
| | - Feng Wei
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, Tianjin, China
| | - Xiubao Ren
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, Tianjin, China.
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, Tianjin, China.
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, Tianjin, China.
- Department of Biotherapy, Tianjin Medical University Cancer Institute and Hospital, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, Tianjin, China.
| |
Collapse
|
6039
|
Bhome R, Al Saihati H, Goh R, Bullock M, Primrose J, Thomas G, Sayan A, Mirnezami A. Translational aspects in targeting the stromal tumour microenvironment: from bench to bedside. NEW HORIZONS IN TRANSLATIONAL MEDICINE 2016; 3:9-21. [PMID: 27275004 PMCID: PMC4888939 DOI: 10.1016/j.nhtm.2016.03.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2016] [Revised: 03/07/2016] [Accepted: 03/24/2016] [Indexed: 02/07/2023]
Abstract
Solid tumours comprise, not only malignant cells but also a variety of stromal cells and extracellular matrix proteins. These components interact via an array of signalling pathways to create an adaptable network that may act to promote or suppress cancer progression. To date, the majority of anti-tumour chemotherapeutic agents have principally sought to target the cancer cell. Consequently, resistance develops because of clonal evolution, as a result of selection pressure during tumour expansion. The concept of activating or inhibiting other cell types within the tumour microenvironment is relatively novel and has the advantage of targeting cells which are genetically stable and less likely to develop resistance. This review outlines key players in the stromal tumour microenvironment and discusses potential targeting strategies that may offer therapeutic benefit.
Collapse
Affiliation(s)
- R. Bhome
- Cancer Sciences, Faculty of Medicine, University of Southampton, Somers Cancer Research Building, Southampton General Hospital, Tremona Road, Southampton SO16 6YD, UK
- University Surgery, South Academic Block, Southampton General Hospital, Tremona Road, Southampton SO16 6YD, UK
| | - H.A. Al Saihati
- Cancer Sciences, Faculty of Medicine, University of Southampton, Somers Cancer Research Building, Southampton General Hospital, Tremona Road, Southampton SO16 6YD, UK
| | - R.W. Goh
- Cancer Sciences, Faculty of Medicine, University of Southampton, Somers Cancer Research Building, Southampton General Hospital, Tremona Road, Southampton SO16 6YD, UK
- School of Medicine, University of Southampton, University Road, Southampton SO17 1BJ, UK
| | - M.D. Bullock
- Cancer Sciences, Faculty of Medicine, University of Southampton, Somers Cancer Research Building, Southampton General Hospital, Tremona Road, Southampton SO16 6YD, UK
- University Surgery, South Academic Block, Southampton General Hospital, Tremona Road, Southampton SO16 6YD, UK
| | - J.N. Primrose
- University Surgery, South Academic Block, Southampton General Hospital, Tremona Road, Southampton SO16 6YD, UK
| | - G.J. Thomas
- Cancer Sciences, Faculty of Medicine, University of Southampton, Somers Cancer Research Building, Southampton General Hospital, Tremona Road, Southampton SO16 6YD, UK
| | - A.E. Sayan
- Cancer Sciences, Faculty of Medicine, University of Southampton, Somers Cancer Research Building, Southampton General Hospital, Tremona Road, Southampton SO16 6YD, UK
| | - A.H. Mirnezami
- Cancer Sciences, Faculty of Medicine, University of Southampton, Somers Cancer Research Building, Southampton General Hospital, Tremona Road, Southampton SO16 6YD, UK
- University Surgery, South Academic Block, Southampton General Hospital, Tremona Road, Southampton SO16 6YD, UK
| |
Collapse
|
6040
|
Smith H, Hayes A. The role of regional chemotherapy in the management of extremity soft tissue malignancies. Eur J Surg Oncol 2016; 42:7-17. [DOI: 10.1016/j.ejso.2015.08.165] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Revised: 08/06/2015] [Accepted: 08/15/2015] [Indexed: 10/23/2022] Open
|
6041
|
Xia Y, Jeffrey Medeiros L, Young KH. Signaling pathway and dysregulation of PD1 and its ligands in lymphoid malignancies. BIOCHIMICA ET BIOPHYSICA ACTA 2016; 1865:58-71. [PMID: 26432723 PMCID: PMC4733614 DOI: 10.1016/j.bbcan.2015.09.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Revised: 09/24/2015] [Accepted: 09/26/2015] [Indexed: 12/12/2022]
Abstract
Tumor cells evade immune destruction, at least partially, by upregulating inhibitory signals to limit effector T cell activation. Programmed death 1 (PD-1) is one of the most critical co-inhibitory molecules limiting the T-cell antitumor response. PD-1 and its ligands, PD-L1 and PD-L2, are overexpressed by various types of tumors as well as reactive cells in the tumor microenvironment. A growing body of evidence has shown the clinical efficiency and minimal toxicity of PD-1 pathway inhibitors in patients with solid tumors, but the role of these inhibitors in lymphoid malignancies is much less well studied. In this review, we analyze the pathologic role of the PD-1 pathway in most common lymphoid malignancies and we organize the clinical data from clinical trials of PD-1 pathway inhibitors. Several anti-PD-1 regimens have shown encouraging therapeutic effects in patients with relapsed or refractory Hodgkin lymphoma, follicular lymphoma, and diffuse large B-cell lymphoma. Additional progress is needed to foster an improved understanding of the role of anti-PD-1 therapy in reconstituting antitumor immunity in patients with lymphoid malignancies. Upcoming trials will explore the clinical efficiency of combining PD-1 pathway inhibitors and various agents with diverse mechanisms of action and create more therapeutic possibilities for afflicted patients.
Collapse
Affiliation(s)
- Yi Xia
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - L Jeffrey Medeiros
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ken H Young
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; The University of Texas Graduate School of Biomedical Science, Houston, TX, USA.
| |
Collapse
|
6042
|
Nagaoka T. Recent Advances in Diagnostic Technologies for Melanoma. ADVANCED BIOMEDICAL ENGINEERING 2016. [DOI: 10.14326/abe.5.111] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Affiliation(s)
- Takashi Nagaoka
- Department of Computational System Biology, Faculty of Biology-Oriented Science and Technology, Kindai University
| |
Collapse
|
6043
|
Aspeslagh S, Postel-Vinay S, Rusakiewicz S, Soria JC, Zitvogel L, Marabelle A. Rationale for anti-OX40 cancer immunotherapy. Eur J Cancer 2016; 52:50-66. [DOI: 10.1016/j.ejca.2015.08.021] [Citation(s) in RCA: 249] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 08/22/2015] [Indexed: 02/08/2023]
|
6044
|
Abstract
Melanoma skin cancer is a potentially deadly disease in humans and has remained extremely difficult to treat once it has metastasized. In just the last 10 years, a number of models of melanoma have been developed in the zebrafish that are biologically faithful to the human disease and have already yielded important insights into the fundamental biology of melanoma and offered new potential avenues for treatment. With the diversity and breadth of the molecular genetic tools available in the zebrafish, these melanoma models will continue to be refined and expanded upon to keep pace with the rapidly evolving field of melanoma biology.
Collapse
|
6045
|
Bell RB, Leidner RS, Crittenden MR, Curti BD, Feng Z, Montler R, Gough MJ, Fox BA, Weinberg AD, Urba WJ. OX40 signaling in head and neck squamous cell carcinoma: Overcoming immunosuppression in the tumor microenvironment. Oral Oncol 2016; 52:1-10. [DOI: 10.1016/j.oraloncology.2015.11.009] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Revised: 10/21/2015] [Accepted: 11/09/2015] [Indexed: 12/12/2022]
|
6046
|
Abstract
Lung cancer has not traditionally been viewed as an immune-responsive tumor. However, it is becoming evident that tumor-induced immune suppression is vital to malignant progression. Immunotherapies act by enhancing the patient's innate immune response and hold promise for inducing long-term responses in select patients with non-small cell lung cancer (NSCLC) and small cell lung cancer (SCLC). Immune checkpoint inhibitors, in particular, inhibitors to cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) and programmed death 1 (PD-1) and programmed death receptor ligand 1 (PD-L1) have shown promise in early studies and are currently in clinical trials in both small cell lung cancer and non-small cell lung cancer patients. Two large randomized phase III trials recently demonstrated superior overall survival (OS) in patients treated with anti-PD-1 therapy compared to chemotherapy in the second-line setting.
Collapse
|
6047
|
Schilling B, Schadendorf D. [Editorial]. J Dtsch Dermatol Ges 2015; 14:1-2. [PMID: 26713629 DOI: 10.1111/ddg.12932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
6048
|
Wieder T, Brenner E, Braumüller H, Röcken M. Immuntherapie des Melanoms: Wirksamkeit und Wirkungsmechanismen. J Dtsch Dermatol Ges 2015. [DOI: 10.1111/ddg.110_12819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Thomas Wieder
- Universitätshautklinik; Eberhard Karls Universität; Tübingen
| | - Ellen Brenner
- Universitätshautklinik; Eberhard Karls Universität; Tübingen
| | | | - Martin Röcken
- Universitätshautklinik; Eberhard Karls Universität; Tübingen
| |
Collapse
|
6049
|
Vennepureddy A, Thumallapally N, Motilal Nehru V, Atallah JP, Terjanian T. Novel Drugs and Combination Therapies for the Treatment of Metastatic Melanoma. J Clin Med Res 2015; 8:63-75. [PMID: 26767073 PMCID: PMC4701060 DOI: 10.14740/jocmr2424w] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/07/2015] [Indexed: 12/28/2022] Open
Abstract
Metastatic melanoma (MM) still remains as one of the most worrisome cancer known to mankind. In last two decades, treatment of melanoma took a dramatic turn with the discovery of targeted therapy which targets the mutations in mitogen-activated protein kinase (MAPK) pathway and immune checkpoint inhibitors. These new findings have led to emergence of many novel drugs that have been approved by FDA. Targeted therapy drugs such as vemurafenib, trametinib and dabrafenib target the MAPK pathway whereas immunotherapies such as ipilimumab, nivolumab and pembrolizumab block immune checkpoint receptors on T lymphocytes. All these drugs have shown to improve the overall survival in MM. Despite these recent discoveries, treatment of MM remains challenging because of rapid development of resistance to targeted therapy. This review will discuss recently approved drugs and their adverse effects and also shed light on combination therapy in treatment of melanoma.
Collapse
Affiliation(s)
- Adarsh Vennepureddy
- Department of Medicine, Staten Island University Hospital, Staten Island, NY 10305, USA
| | | | | | - Jean-Paul Atallah
- Division of Hematolgy and Oncology, Staten Island University Hospital, Staten Island, NY 10305, USA
| | - Terenig Terjanian
- Division of Hematolgy and Oncology, Staten Island University Hospital, Staten Island, NY 10305, USA
| |
Collapse
|
6050
|
Champiat S, Lambotte O, Barreau E, Belkhir R, Berdelou A, Carbonnel F, Cauquil C, Chanson P, Collins M, Durrbach A, Ederhy S, Feuillet S, François H, Lazarovici J, Le Pavec J, De Martin E, Mateus C, Michot JM, Samuel D, Soria JC, Robert C, Eggermont A, Marabelle A. Management of immune checkpoint blockade dysimmune toxicities: a collaborative position paper. Ann Oncol 2015; 27:559-74. [PMID: 26715621 DOI: 10.1093/annonc/mdv623] [Citation(s) in RCA: 645] [Impact Index Per Article: 64.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 12/18/2015] [Indexed: 12/13/2022] Open
Abstract
Monoclonal antibodies targeted against the immune checkpoint molecules CTLA-4 and PD-1 have recently obtained approval for the treatment of metastatic melanoma and advanced/refractory non small-cell lung cancers. Therefore, their use will not be limited anymore to selected hospitals involved in clinical trials. Indeed, they will be routinely prescribed in many cancer centers across the world. Besides their efficacy profile, these immune targeted agents also generate immune-related adverse events (irAEs). This new family of dysimmune toxicities remains largely unknown to the broad oncology community. Although severe irAEs remain rare (∼10% of cases under monotherapy), they can become life-threatening if not anticipated and managed appropriately. Over the last 5 years, Gustave Roussy has accumulated a significant experience in the prescription of immune checkpoint blockade (ICB) antibodies and the management of their toxicities. Together with the collaboration of Gustave Roussy's network of organ specialists with expertise in irAEs, we propose here some practical guidelines for the oncologist to help in the clinical care of patients under ICB immunotherapy.
Collapse
Affiliation(s)
- S Champiat
- Department of Drug Development (DITEP), Gustave Roussy Inserm U981, Univ. Paris-Sud, Université Paris-Saclay, Villejuif
| | - O Lambotte
- Department of Internal Medicine and Clinical Immunology, Assistance Publique-Hôpitaux de Paris, Hôpital Universitaire Bicêtre, Le Kremlin Bicêtre Université Paris Sud 11, Le Kremlin-Bicêtre CEA, DSV/iMETI, Division of Immuno-Virology, IDMIT, Fontenay-aux-Roses INSERM, U1184, Center for Immunology of Viral Infections and Autoimmune Diseases, Le Kremlin-Bicêtre
| | - E Barreau
- Department of Ophthalmology, Hôpital Universitaire Bicêtre, Le Kremlin Bicêtre
| | - R Belkhir
- Department of Rheumatology, Hôpitaux Universitaires Paris-Sud, Hôpital Bicêtre, AP-HP, Le Kremlin Bicêtre
| | - A Berdelou
- Department of Nuclear Medicine and Endocrine Tumors, Gustave Roussy, Univ. Paris-Sud, Université Paris-Saclay, Villejuif
| | - F Carbonnel
- Gastroenterology Unit, Université Paris-Sud, Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Paris Sud, Le Kremlin Bicêtre
| | - C Cauquil
- Division of Adult Neurology, Hôpital Universitaire Bicêtre, Le Kremlin Bicêtre
| | - P Chanson
- Faculty of Medicine, Université Paris-Saclay, Univ Paris-Sud, Paris-Sud, UMR-S1185, Le Kremlin Bicêtre Unit of Endocrinology and reproductive Health, Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Paris-Sud, Hôpital de Bicêtre, Le Kremlin Bicêtre Unit of Gastroenterology, Institut National de la Santé et de la Recherche Médicale U1185 (P.C.), Le Kremlin Bicêtre
| | - M Collins
- Gastroenterology Unit, Université Paris-Sud, Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Paris Sud, Le Kremlin Bicêtre
| | - A Durrbach
- Department of Nephrology and Transplantation, Bicêtre Hospital, Paris Saclay University, INSERM 1197, Le Kremlin Bicêtre
| | - S Ederhy
- Department of Cardiology, Saint-Antoine Hospital, Assistance Publique-Hôpitaux de Paris, Pierre et Marie Curie University [UPMC], Paris-Sorbonne, Paris
| | - S Feuillet
- Department of Thoracic and cardiovascular, and transplantation cardio-pulmonary, Hôpital Marie-Lannelongue, Le Plessis-Robinson Univ. Paris-Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin Bicêtre
| | - H François
- Department of Nephrology and Transplantation, Bicêtre Hospital, Paris Saclay University, INSERM 1197, Le Kremlin Bicêtre
| | - J Lazarovici
- Hematology Unit, Department of Medical Oncology, Gustave Roussy, Univ. Paris-Sud, Université Paris-Saclay, Villejuif
| | - J Le Pavec
- Department of Thoracic and cardiovascular, and transplantation cardio-pulmonary, Hôpital Marie-Lannelongue, Le Plessis-Robinson Univ. Paris-Sud, Faculté de Médecine, Université Paris-Saclay, Le Kremlin Bicêtre UMR_S 999, Univ. Paris-Sud; INSERM; Hôpital Marie Lannelongue, Le Plessis Robinson
| | - E De Martin
- Centre Hépato-Biliaire, AP-HP, Hôpital Universitaire Paul Brousse Inserm U1193
| | - C Mateus
- Dermatology Unit, Department of Medical Oncology, Gustave Roussy, Univ. Paris-Sud, Université Paris-Saclay, Villejuif
| | - J-M Michot
- Department of Drug Development (DITEP), Gustave Roussy
| | - D Samuel
- Centre Hépato-Biliaire, AP-HP, Hôpital Universitaire Paul Brousse Inserm U1193
| | - J-C Soria
- Department of Drug Development (DITEP), Gustave Roussy Inserm U981, Univ. Paris-Sud, Université Paris-Saclay, Villejuif
| | - C Robert
- Inserm U981, Univ. Paris-Sud, Université Paris-Saclay, Villejuif Dermatology Unit, Department of Medical Oncology, Gustave Roussy, Univ. Paris-Sud, Université Paris-Saclay, Villejuif
| | | | - A Marabelle
- Department of Drug Development (DITEP), Gustave Roussy Gustave Roussy Cancer Campus, Villejuif Inserm 1015, Univ. Paris-Sud, Université Paris-Saclay, Villejuif, France
| |
Collapse
|