5501
|
Bansal P, Osman D, Gan GN, Simon GR, Boumber Y. Recent Advances in Immunotherapy in Metastatic NSCLC. Front Oncol 2016; 6:239. [PMID: 27896216 PMCID: PMC5107578 DOI: 10.3389/fonc.2016.00239] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2016] [Accepted: 10/26/2016] [Indexed: 12/22/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) is one of most common malignancies and the leading cause of cancer deaths worldwide. Despite advances in targeted therapies, majority of NSCLC patients do not have targetable genomic alterations. Nevertheless, recent discovery that NSCLC is an immunogenic tumor type, and several breakthroughs in immunotherapies have led to rapid expansion of this new treatment modality in NSCLC with recent FDA approvals of programed death receptor-1 inhibitors, such as nivolumab and pembrolizumab. Here, we review promising immunotherapeutic approaches in metastatic NSCLC, including checkpoint inhibitors, agents with other mechanisms of action, and immunotherapy combinations with other drugs. With advent of immunotherapy, therapeutic options in metastatic NSCLC are rapidly expanding with the hope to further expand life expectancy in metastatic lung cancer.
Collapse
Affiliation(s)
- Pranshu Bansal
- Department of Internal Medicine, Division of Hematology/Oncology, University of New Mexico Comprehensive Cancer Center, University of New Mexico School of Medicine, Albuquerque, NM, USA; Hematology/Oncology Fellowship Program, University of New Mexico Comprehensive Cancer Center, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Diaa Osman
- Department of Internal Medicine, Division of Hematology/Oncology, University of New Mexico Comprehensive Cancer Center, University of New Mexico School of Medicine, Albuquerque, NM, USA; Hematology/Oncology Fellowship Program, University of New Mexico Comprehensive Cancer Center, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Gregory N Gan
- Department of Internal Medicine, Division of Hematology/Oncology, University of New Mexico Comprehensive Cancer Center, University of New Mexico School of Medicine, Albuquerque, NM, USA; Section of Radiation Oncology, University of New Mexico Comprehensive Cancer Center, Albuquerque, NM, USA
| | - George R Simon
- Department of Thoracic and Head/Neck Medical Oncology, Division of Cancer Medicine, University of Texas MD Anderson Cancer Center , Houston, TX , USA
| | - Yanis Boumber
- Department of Internal Medicine, Division of Hematology/Oncology, University of New Mexico Comprehensive Cancer Center, University of New Mexico School of Medicine, Albuquerque, NM, USA; Molecular Therapeutics Program, Department of Hematology/Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA
| |
Collapse
|
5502
|
The Future Prospects of Immune Therapy in Gastric and Esophageal Adenocarcinoma. J Clin Med 2016; 5:jcm5110100. [PMID: 27854242 PMCID: PMC5126797 DOI: 10.3390/jcm5110100] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 11/02/2016] [Accepted: 11/02/2016] [Indexed: 12/12/2022] Open
Abstract
The prognosis of esophageal cancers is poor and novel approaches are urgently needed. Despite improvements in outcomes with transtuzumab and ramucirumab, these improvements added an average of only 2 to 3 months with a median overall survival reported to be around 1 year. Comprehensive genomic sequencing has defined some molecular alterations with potential targets, but the majority of patients still do not benefit from druggable targets. Breakthroughs in immune checkpoint blockade have provided new therapeutic options in many cancers. Programmed death ligand 1 (PDL1) overexpression, a possible biomarker predicting response to immune checkpoint inhibitors, approaches forty percent in esophageal and gastric cancers. Translational and molecular studies have shown that esophageal cancers are possible candidate malignancies for immune checkpoint inhibition. In this review, we plan to highlight the mechanisms, preclinical, and early clinical data that provide insight on the role of immune therapeutics in esophageal cancers.
Collapse
|
5503
|
Affiliation(s)
- Won Kim
- Won Kim and Lawrence Fong, University of California, San Francisco, San Francisco, CA
| | - Lawrence Fong
- Won Kim and Lawrence Fong, University of California, San Francisco, San Francisco, CA
| |
Collapse
|
5504
|
Tripp MK, Watson M, Balk SJ, Swetter SM, Gershenwald JE. State of the science on prevention and screening to reduce melanoma incidence and mortality: The time is now. CA Cancer J Clin 2016; 66:460-480. [PMID: 27232110 PMCID: PMC5124531 DOI: 10.3322/caac.21352] [Citation(s) in RCA: 128] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Answer questions and earn CME/CNE Although overall cancer incidence rates are decreasing, melanoma incidence rates continue to increase about 3% annually. Melanoma is a significant public health problem that exacts a substantial financial burden. Years of potential life lost from melanoma deaths contribute to the social, economic, and human toll of this disease. However, most cases are potentially preventable. Research has clearly established that exposure to ultraviolet radiation increases melanoma risk. Unprecedented antitumor activity and evolving survival benefit from novel targeted therapies and immunotherapies are now available for patients with unresectable and/or metastatic melanoma. Still, prevention (minimizing sun exposure that may result in tanned or sunburned skin and avoiding indoor tanning) and early detection (identifying lesions before they become invasive or at an earlier stage) have significant potential to reduce melanoma incidence and melanoma-associated deaths. This article reviews the state of the science on prevention and early detection of melanoma and current areas of scientific uncertainty and ongoing debate. The US Surgeon General's Call to Action to Prevent Skin Cancer and US Preventive Services Task Force reviews on skin cancer have propelled a national discussion on melanoma prevention and screening that makes this an extraordinary and exciting time for diverse disciplines in multiple sectors-health care, government, education, business, advocacy, and community-to coordinate efforts and leverage existing knowledge to make major strides in reducing the public health burden of melanoma in the United States. CA Cancer J Clin 2016;66:460-480. © 2016 American Cancer Society.
Collapse
Affiliation(s)
- Mary K Tripp
- Instructor, Department of Behavioral Science, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Meg Watson
- Epidemiologist, Division of Cancer Prevention and Control, Centers for Disease Control and Prevention, Atlanta, GA
| | - Sophie J Balk
- Attending Pediatrician, Children's Hospital at Montefiore, and Professor of Clinical Pediatrics, Albert Einstein College of Medicine, Bronx, NY
| | - Susan M Swetter
- Professor, Department of Dermatology, and Director, Pigmented Lesion and Melanoma Program, Stanford University Medical Center and Cancer Institute, Stanford, CA
- Professor and Assistant Chief, Dermatology Service, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA
| | - Jeffrey E Gershenwald
- Dr. John M. Skibber Professor, Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
- Professor, Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX
- Medical Director, Melanoma and Skin Center, The University of Texas MD Anderson Cancer Center, Houston, TX
- Co-Leader, Melanoma Moon Shot, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
5505
|
Sun TY, Haberman AM, Greco V. Preclinical Advances with Multiphoton Microscopy in Live Imaging of Skin Cancers. J Invest Dermatol 2016; 137:282-287. [PMID: 27847119 DOI: 10.1016/j.jid.2016.08.033] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 08/09/2016] [Accepted: 08/24/2016] [Indexed: 01/13/2023]
Abstract
Conventional, static analyses have historically been the bedrock and tool of choice for the study of skin cancers. Over the past several years, in vivo imaging of tumors using multiphoton microscopy has emerged as a powerful preclinical tool for revealing detailed cellular behaviors from the earliest moments of tumor development to the final steps of metastasis. Multiphoton microscopy allows for deep tissue penetration with relatively minor phototoxicity, rendering it an effective tool for the long-term observation of tumor evolution. This review highlights some of the recent preclinical insights gained using multiphoton microscopy and suggests future advances that could enhance its power in revealing the mysteries of skin tumor biology.
Collapse
Affiliation(s)
- Thomas Yang Sun
- Department of Genetics, Yale School of Medicine, New Haven, Connecticut, USA.
| | - Ann M Haberman
- Departments of Immunobiology and Laboratory Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Valentina Greco
- Department of Genetics, Yale School of Medicine, New Haven, Connecticut, USA; Departments of Dermatology and Cell Biology, Yale Stem Cell Center, Yale Cancer Center, Yale School of Medicine, New Haven, Connecticut, USA.
| |
Collapse
|
5506
|
Combinatorial immunotherapy for melanoma. Cancer Gene Ther 2016; 24:141-147. [PMID: 27834353 DOI: 10.1038/cgt.2016.56] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 09/29/2016] [Indexed: 12/18/2022]
Abstract
Melanoma has been a long-standing focal point for immunotherapy development. In this review, we explore the evolution of melanoma treatments with particular attention to the history and recent advances in melanoma immunotherapy. We also discuss novel combinations of these modalities and their potential to offer novel therapeutic options for patients with advanced melanoma.
Collapse
|
5507
|
Jamieson NB, Maker AV. Gene-expression profiling to predict responsiveness to immunotherapy. Cancer Gene Ther 2016; 24:134-140. [PMID: 27834354 PMCID: PMC5386795 DOI: 10.1038/cgt.2016.63] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2016] [Accepted: 10/06/2016] [Indexed: 12/17/2022]
Abstract
Recent clinical successes with immunotherapy have resulted in expanding indications for cancer therapy. To enhance anti-tumor immune responses, and to better choose specific strategies matched to patient and tumor characteristics, genomic-driven precision immunotherapy will be necessary. Herein, we explore the role that tumor gene expression profiling (GEP) and transcriptome expression may play in the prediction of an immunotherapeutic response. Genetic markers associated with response to immunotherapy are addressed as they pertain to the tumor genomic landscape, the extent of DNA damage, tumor mutational load, and tumor-specific neoantigens. Furthermore, genetic markers associated with resistance to checkpoint blockade and relapse are reviewed. Finally, the utility of GEP to identify new tumor types for immunotherapy and implications for combinatorial strategies are summarized.
Collapse
Affiliation(s)
- N B Jamieson
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences and the Academic Unit of Surgery, School of Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, Scotland.,West of Scotland Pancreatic Unit, Glasgow Royal Infirmary, Glasgow, Scotland
| | - A V Maker
- Department of Surgery, Division of Surgical Oncology; Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|
5508
|
Density of immunogenic antigens does not explain the presence or absence of the T-cell-inflamed tumor microenvironment in melanoma. Proc Natl Acad Sci U S A 2016; 113:E7759-E7768. [PMID: 27837020 DOI: 10.1073/pnas.1609376113] [Citation(s) in RCA: 313] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Melanoma metastases can be categorized by gene expression for the presence of a T-cell-inflamed tumor microenvironment, which correlates with clinical efficacy of immunotherapies. T cells frequently recognize mutational antigens corresponding to nonsynonymous somatic mutations (NSSMs), and in some cases shared differentiation or cancer-testis antigens. Therapies are being pursued to trigger immune infiltration into non-T-cell-inflamed tumors in the hope of rendering them immunotherapy responsive. However, whether those tumors express antigens capable of T-cell recognition has not been explored. To address this question, 266 melanomas from The Cancer Genome Atlas (TCGA) were categorized by the presence or absence of a T-cell-inflamed gene signature. These two subsets were interrogated for cancer-testis, differentiation, and somatic mutational antigens. No statistically significant differences were observed, including density of NSSMs. Focusing on hypothetical HLA-A2+ binding scores, 707 peptides were synthesized, corresponding to all identified candidate neoepitopes. No differences were observed in measured HLA-A2 binding between inflamed and noninflamed cohorts. Twenty peptides were randomly selected from each cohort to evaluate priming and recognition by human CD8+ T cells in vitro with 25% of peptides confirmed to be immunogenic in both. A similar gene expression profile applied to all solid tumors of TCGA revealed no association between T-cell signature and NSSMs. Our results indicate that lack of spontaneous immune infiltration in solid tumors is unlikely due to lack of antigens. Strategies that improve T-cell infiltration into tumors may therefore be able to facilitate clinical response to immunotherapy once antigens become recognized.
Collapse
|
5509
|
Ring EK, Markert JM, Gillespie GY, Friedman GK. Checkpoint Proteins in Pediatric Brain and Extracranial Solid Tumors: Opportunities for Immunotherapy. Clin Cancer Res 2016; 23:342-350. [PMID: 27836863 DOI: 10.1158/1078-0432.ccr-16-1829] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 09/12/2016] [Accepted: 10/10/2016] [Indexed: 12/31/2022]
Abstract
Pediatric brain and extracranial solid tumors are a diverse group of malignancies that represent almost half of all pediatric cancers. Standard therapy includes various combinations of surgery, cytotoxic chemotherapy, and radiation, which can be very harmful to a developing child, and survivors carry a substantial burden of long-term morbidities. Although these therapies have improved survival rates for children with solid tumors, outcomes still remain extremely poor for subsets of patients. Recently, immunosuppressive checkpoint molecules that negatively regulate immune cell function have been described. When found on malignant cells or in the tumor microenvironment, they contribute to immune evasion and tumor escape. Agents designed to inhibit these proteins have demonstrated significant efficacy in human adult solid tumor studies. However, there is limited research focusing on immune checkpoint molecules and inhibitors in pediatric solid tumors. In this review, we examine the current knowledge on immune checkpoint proteins with an emphasis on cytotoxic T lymphocyte antigen-4 (CTLA-4); programmed cell death protein-1 (PD-1) and programmed death-ligand 1 (PD-L1); OX-2 membrane glycoprotein (CD200); and indoleamine 2,3-dioxygenase (IDO). We review T-cell signaling, the mechanisms of action of these checkpoint molecules, pediatric preclinical studies on checkpoint proteins and checkpoint blockade, pediatric checkpoint inhibitor clinical trials conducted to date, and future immunotherapy opportunities for childhood cancers. Clin Cancer Res; 23(2); 342-50. ©2016 AACR.
Collapse
Affiliation(s)
- Eric K Ring
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, Alabama
| | - James M Markert
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, Alabama
| | - G Yancey Gillespie
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, Alabama
| | - Gregory K Friedman
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, Alabama.
| |
Collapse
|
5510
|
McLachlan J, Boussios S, Okines A, Glaessgen D, Bodlar S, Kalaitzaki R, Taylor A, Lalondrelle S, Gore M, Kaye S, Banerjee S. The Impact of Systemic Therapy Beyond First-line Treatment for Advanced Cervical Cancer. Clin Oncol (R Coll Radiol) 2016; 29:153-160. [PMID: 27838135 DOI: 10.1016/j.clon.2016.10.002] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 10/03/2016] [Accepted: 10/06/2016] [Indexed: 10/20/2022]
Abstract
AIMS Despite recent advances in the primary and secondary prevention of cervical cancer, a significant number of women present with or develop metastatic disease. There is currently no consensus on the standard of care for second-line systemic treatment of recurrent/metastatic cervical cancer. The purpose of this study was to evaluate the second-line systemic therapy used and the associated outcomes in a single cancer centre. MATERIALS AND METHODS A retrospective review of patients with cervical cancer who received one or more lines of treatment for recurrent or metastatic cervical cancer at the Royal Marsden Hospital between 2004 and 2014 was carried out. The primary objective was to establish the types of second-line systemic treatment used. Secondary end points included objective response rate, progression-free survival and overall survival after second-line therapy. RESULTS In total, 75 patients were included in the study; 53 patients (70.7%) received second-line therapy for recurrent/metastatic disease. The most common second-line therapy was weekly paclitaxel (28.3%). Carboplatin-based chemotherapy (24.5%), targeted agent monotherapy within clinical trials (22.6%), docetaxel-based chemotherapy (13.2%), topotecan (9.4%) and gemcitabine (1.9%) were also used. The objective response rate to second-line therapy was 13.2%, which included three partial responses to carboplatin and paclitaxel, two partial responses to docetaxel-based chemotherapy, one partial response to weekly paclitaxel and one partial response to cediranib. Twenty-two patients (41.5%) achieved stable disease at 4 months. The median progression-free survival for women treated with second-line therapy was 3.2 months (95% confidence interval 2.1-4.3) and median overall survival was 9.3 months (95% confidence interval 6.4-12.5). Thirty-nine per cent of patients received third-line therapy. CONCLUSION Seventy per cent of patients treated with first-line systemic therapy for recurrent/metastatic cervical cancer subsequently received second-line treatment but response rates were poor. There remains no standard of care for second-line systemic therapy for advanced cervical cancer. Patients should be considered for clinical trials whenever feasible, including novel targeted agents and immunotherapy.
Collapse
Affiliation(s)
- J McLachlan
- Gynaecology Unit, The Royal Marsden NHS Foundation Trust, London, UK
| | - S Boussios
- Gynaecology Unit, The Royal Marsden NHS Foundation Trust, London, UK
| | - A Okines
- Gynaecology Unit, The Royal Marsden NHS Foundation Trust, London, UK
| | - D Glaessgen
- Gynaecology Unit, The Royal Marsden NHS Foundation Trust, London, UK
| | - S Bodlar
- Research Data and Statistics Unit, Royal Marsden NHS Foundation Trust, London, UK
| | - R Kalaitzaki
- Research Data and Statistics Unit, Royal Marsden NHS Foundation Trust, London, UK
| | - A Taylor
- Gynaecology Unit, The Royal Marsden NHS Foundation Trust, London, UK
| | - S Lalondrelle
- Gynaecology Unit, The Royal Marsden NHS Foundation Trust, London, UK
| | - M Gore
- Gynaecology Unit, The Royal Marsden NHS Foundation Trust, London, UK
| | - S Kaye
- Gynaecology Unit, The Royal Marsden NHS Foundation Trust, London, UK
| | - S Banerjee
- Gynaecology Unit, The Royal Marsden NHS Foundation Trust and Institute of Cancer Research, London, UK.
| |
Collapse
|
5511
|
Making urothelial carcinomas less immune to immunotherapy. Urol Oncol 2016; 34:534-537. [PMID: 27836245 DOI: 10.1016/j.urolonc.2016.10.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 10/05/2016] [Indexed: 12/26/2022]
Abstract
The success of immune checkpoint inhibitors in advanced urothelial carcinoma provides patients with the prospect for durable objective responses. However, the majority of patients do not respond to immune checkpoint blockade. Several potential predictive biomarkers of response have been evaluated in hopes of better identifying likely responders, though each has been shown to have limitations. Going forward, development of reliable predictive biomarkers is imperative. Likewise, innovative treatment combination approaches to convert non-responders to responders are essential to continue making progress in the field.
Collapse
|
5512
|
Jelinek T, Hajek R. PD-1/PD-L1 inhibitors in multiple myeloma: The present and the future. Oncoimmunology 2016; 5:e1254856. [PMID: 28123899 DOI: 10.1080/2162402x.2016.1254856] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 10/22/2016] [Accepted: 10/24/2016] [Indexed: 01/05/2023] Open
Abstract
The introduction of PD-1/PD-L1 pathway inhibitors has marked a significant milestone in the treatment of various types of solid tumors. The current situation in multiple myeloma (MM) is rather unclear, as distinct research groups have reported discordant results. This discrepancy dominantly concerns the expression of PD-1/PD-L1 molecules as well as the identification of the responsible immune effector cell population. The results of monotherapy with PD-1/PD-L1 inhibitors have been unsatisfactory in MM, suggesting that a combination approach is needed. The most logical partners are immunomodulatory agents as they possess many synergistic effects. We are also proposing other rational and promising combinations (e.g., daratumumab, ibrutinib, anti-CD137) that warrant further investigation.
Collapse
Affiliation(s)
- T Jelinek
- Faculty of Science, University of Ostrava, Czech Republic; Department of Haematooncology, University Hospital Ostrava and Faculty of Medicine, University of Ostrava, Czech Republic; Centro de Investigacion Medica Aplicada (CIMA), Clinica Universidad de Navarra, IDISNA, Pamplona, Spain
| | - R Hajek
- Department of Haematooncology, University Hospital Ostrava and Faculty of Medicine, University of Ostrava , Czech Republic
| |
Collapse
|
5513
|
Abstract
INTRODUCTION The treatment of melanoma is evolving rapidly over the past few years. Areas covered: We conducted a comprehensive review of the literature on the role of nivolumab in melanoma Expert commentary: Nivolumab is approved by FDA and EMA for the treatment of patients with metastatic melanoma. Nivolumab is superior to chemotherapy and to ipilimumab in previously untreated patients and to chemotherapy in ipilimumab pre-treated patients. The addition ipilimumab to nivolumab is associated with a higher response rate and a better PFS, particularly in patients with PD-L1 negative tumors, albeit at the cost of an increase in grade 3-4 adverse event rate. Definitive survival data on this combination are pending and the selection of patients most likely to benefit from this combination and its pharmacoeconomics are to be elucidated. Prospectively validated predictive markers are lacking. Of particular interest are immune-related adverse events which should be managed according to published guidelines.
Collapse
Affiliation(s)
- Pol Specenier
- a Faculty of Medicine and Health Sciences , University of Antwerp , Antwerp , Belgium.,b Department of Medical Oncology , Antwerp University Hospital , Edegem , Belgium
| |
Collapse
|
5514
|
D'Angelo SP, Larkin J, Sosman JA, Lebbé C, Brady B, Neyns B, Schmidt H, Hassel JC, Hodi FS, Lorigan P, Savage KJ, Miller WH, Mohr P, Marquez-Rodas I, Charles J, Kaatz M, Sznol M, Weber JS, Shoushtari AN, Ruisi M, Jiang J, Wolchok JD. Efficacy and Safety of Nivolumab Alone or in Combination With Ipilimumab in Patients With Mucosal Melanoma: A Pooled Analysis. J Clin Oncol 2016; 35:226-235. [PMID: 28056206 DOI: 10.1200/jco.2016.67.9258] [Citation(s) in RCA: 433] [Impact Index Per Article: 48.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Purpose Mucosal melanoma is an aggressive malignancy with a poor response to conventional therapies. The efficacy and safety of nivolumab (a programmed death-1 checkpoint inhibitor), alone or combined with ipilimumab (a cytotoxic T-lymphocyte antigen-4 checkpoint inhibitor), have not been reported in this rare melanoma subtype. Patients and Methods Data were pooled from 889 patients who received nivolumab monotherapy in clinical studies, including phase III trials; 86 (10%) had mucosal melanoma and 665 (75%) had cutaneous melanoma. Data were also pooled for patients who received nivolumab combined with ipilimumab (n = 35, mucosal melanoma; n = 326, cutaneous melanoma). Results Among patients who received nivolumab monotherapy, median progression-free survival was 3.0 months (95% CI, 2.2 to 5.4 months) and 6.2 months (95% CI, 5.1 to 7.5 months) for mucosal and cutaneous melanoma, with objective response rates of 23.3% (95% CI, 14.8% to 33.6%) and 40.9% (95% CI, 37.1% to 44.7%), respectively. Median progression-free survival in patients treated with nivolumab combined with ipilimumab was 5.9 months (95% CI, 2.8 months to not reached) and 11.7 months (95% CI, 8.9 to 16.7 months) for mucosal and cutaneous melanoma, with objective response rates of 37.1% (95% CI, 21.5% to 55.1%) and 60.4% (95% CI, 54.9% to 65.8%), respectively. For mucosal and cutaneous melanoma, respectively, the incidence of grade 3 or 4 treatment-related adverse events was 8.1% and 12.5% for nivolumab monotherapy and 40.0% and 54.9% for combination therapy. Conclusion To our knowledge, this is the largest analysis of data for anti-programmed death-1 therapy in mucosal melanoma to date. Nivolumab combined with ipilimumab seemed to have greater efficacy than either agent alone, and although the activity was lower in mucosal melanoma, the safety profile was similar between subtypes.
Collapse
Affiliation(s)
- Sandra P D'Angelo
- Sandra P. D'Angelo, Alexander N. Shoushtari, and Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY; James Larkin, Royal Marsden Hospital, London; Paul Lorigan, University of Manchester, Manchester, United Kingdom; Jeffrey A. Sosman, Vanderbilt University Medical Center, Nashville, TN; Celeste Lebbé, Saint-Louis Hospital, Institut National de la Santé et de la Recherche Médicale U976, Université Paris Diderot, Paris; Julie Charles, Grenoble University Hospital, Grenoble Alps University, Grenoble, France; Benjamin Brady, Cabrini Health, Melbourne, Australia; Bart Neyns, Universitair Ziekenhuis Brussel, Brussels, Belgium; Henrik Schmidt, Århus University, Åarhus, Denmark; Jessica C. Hassel, University Hospital Heidelberg, Heidelberg; Peter Mohr, Elbe Kliniken Buxtehude, Buxtehude; Martin Kaatz, SRH Waldklinikum Gera, University Hospital Jena, Jena, Germany; F. Stephen Hodi, Dana-Farber Cancer Institute, Boston, MA; Kerry J. Savage, BC Cancer Agency, University of British Columbia, Vancouver; Wilson H. Miller Jr, Lady Davis Institute and Jewish General Hospital, McGill University, Montreal, Canada; Ivan Marquez-Rodas, Hospital General Universitario Gregorio Marañón, Madrid, Spain; Mario Sznol, Yale University School of Medicine and Smilow Cancer Center, Yale-New Haven Hospital, New Haven, CT; Jeffrey S. Weber, Moffitt Cancer Center, Tampa, FL; and Mary Ruisi and Joel Jiang, Bristol-Myers Squibb, Princeton, NJ
| | - James Larkin
- Sandra P. D'Angelo, Alexander N. Shoushtari, and Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY; James Larkin, Royal Marsden Hospital, London; Paul Lorigan, University of Manchester, Manchester, United Kingdom; Jeffrey A. Sosman, Vanderbilt University Medical Center, Nashville, TN; Celeste Lebbé, Saint-Louis Hospital, Institut National de la Santé et de la Recherche Médicale U976, Université Paris Diderot, Paris; Julie Charles, Grenoble University Hospital, Grenoble Alps University, Grenoble, France; Benjamin Brady, Cabrini Health, Melbourne, Australia; Bart Neyns, Universitair Ziekenhuis Brussel, Brussels, Belgium; Henrik Schmidt, Århus University, Åarhus, Denmark; Jessica C. Hassel, University Hospital Heidelberg, Heidelberg; Peter Mohr, Elbe Kliniken Buxtehude, Buxtehude; Martin Kaatz, SRH Waldklinikum Gera, University Hospital Jena, Jena, Germany; F. Stephen Hodi, Dana-Farber Cancer Institute, Boston, MA; Kerry J. Savage, BC Cancer Agency, University of British Columbia, Vancouver; Wilson H. Miller Jr, Lady Davis Institute and Jewish General Hospital, McGill University, Montreal, Canada; Ivan Marquez-Rodas, Hospital General Universitario Gregorio Marañón, Madrid, Spain; Mario Sznol, Yale University School of Medicine and Smilow Cancer Center, Yale-New Haven Hospital, New Haven, CT; Jeffrey S. Weber, Moffitt Cancer Center, Tampa, FL; and Mary Ruisi and Joel Jiang, Bristol-Myers Squibb, Princeton, NJ
| | - Jeffrey A Sosman
- Sandra P. D'Angelo, Alexander N. Shoushtari, and Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY; James Larkin, Royal Marsden Hospital, London; Paul Lorigan, University of Manchester, Manchester, United Kingdom; Jeffrey A. Sosman, Vanderbilt University Medical Center, Nashville, TN; Celeste Lebbé, Saint-Louis Hospital, Institut National de la Santé et de la Recherche Médicale U976, Université Paris Diderot, Paris; Julie Charles, Grenoble University Hospital, Grenoble Alps University, Grenoble, France; Benjamin Brady, Cabrini Health, Melbourne, Australia; Bart Neyns, Universitair Ziekenhuis Brussel, Brussels, Belgium; Henrik Schmidt, Århus University, Åarhus, Denmark; Jessica C. Hassel, University Hospital Heidelberg, Heidelberg; Peter Mohr, Elbe Kliniken Buxtehude, Buxtehude; Martin Kaatz, SRH Waldklinikum Gera, University Hospital Jena, Jena, Germany; F. Stephen Hodi, Dana-Farber Cancer Institute, Boston, MA; Kerry J. Savage, BC Cancer Agency, University of British Columbia, Vancouver; Wilson H. Miller Jr, Lady Davis Institute and Jewish General Hospital, McGill University, Montreal, Canada; Ivan Marquez-Rodas, Hospital General Universitario Gregorio Marañón, Madrid, Spain; Mario Sznol, Yale University School of Medicine and Smilow Cancer Center, Yale-New Haven Hospital, New Haven, CT; Jeffrey S. Weber, Moffitt Cancer Center, Tampa, FL; and Mary Ruisi and Joel Jiang, Bristol-Myers Squibb, Princeton, NJ
| | - Celeste Lebbé
- Sandra P. D'Angelo, Alexander N. Shoushtari, and Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY; James Larkin, Royal Marsden Hospital, London; Paul Lorigan, University of Manchester, Manchester, United Kingdom; Jeffrey A. Sosman, Vanderbilt University Medical Center, Nashville, TN; Celeste Lebbé, Saint-Louis Hospital, Institut National de la Santé et de la Recherche Médicale U976, Université Paris Diderot, Paris; Julie Charles, Grenoble University Hospital, Grenoble Alps University, Grenoble, France; Benjamin Brady, Cabrini Health, Melbourne, Australia; Bart Neyns, Universitair Ziekenhuis Brussel, Brussels, Belgium; Henrik Schmidt, Århus University, Åarhus, Denmark; Jessica C. Hassel, University Hospital Heidelberg, Heidelberg; Peter Mohr, Elbe Kliniken Buxtehude, Buxtehude; Martin Kaatz, SRH Waldklinikum Gera, University Hospital Jena, Jena, Germany; F. Stephen Hodi, Dana-Farber Cancer Institute, Boston, MA; Kerry J. Savage, BC Cancer Agency, University of British Columbia, Vancouver; Wilson H. Miller Jr, Lady Davis Institute and Jewish General Hospital, McGill University, Montreal, Canada; Ivan Marquez-Rodas, Hospital General Universitario Gregorio Marañón, Madrid, Spain; Mario Sznol, Yale University School of Medicine and Smilow Cancer Center, Yale-New Haven Hospital, New Haven, CT; Jeffrey S. Weber, Moffitt Cancer Center, Tampa, FL; and Mary Ruisi and Joel Jiang, Bristol-Myers Squibb, Princeton, NJ
| | - Benjamin Brady
- Sandra P. D'Angelo, Alexander N. Shoushtari, and Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY; James Larkin, Royal Marsden Hospital, London; Paul Lorigan, University of Manchester, Manchester, United Kingdom; Jeffrey A. Sosman, Vanderbilt University Medical Center, Nashville, TN; Celeste Lebbé, Saint-Louis Hospital, Institut National de la Santé et de la Recherche Médicale U976, Université Paris Diderot, Paris; Julie Charles, Grenoble University Hospital, Grenoble Alps University, Grenoble, France; Benjamin Brady, Cabrini Health, Melbourne, Australia; Bart Neyns, Universitair Ziekenhuis Brussel, Brussels, Belgium; Henrik Schmidt, Århus University, Åarhus, Denmark; Jessica C. Hassel, University Hospital Heidelberg, Heidelberg; Peter Mohr, Elbe Kliniken Buxtehude, Buxtehude; Martin Kaatz, SRH Waldklinikum Gera, University Hospital Jena, Jena, Germany; F. Stephen Hodi, Dana-Farber Cancer Institute, Boston, MA; Kerry J. Savage, BC Cancer Agency, University of British Columbia, Vancouver; Wilson H. Miller Jr, Lady Davis Institute and Jewish General Hospital, McGill University, Montreal, Canada; Ivan Marquez-Rodas, Hospital General Universitario Gregorio Marañón, Madrid, Spain; Mario Sznol, Yale University School of Medicine and Smilow Cancer Center, Yale-New Haven Hospital, New Haven, CT; Jeffrey S. Weber, Moffitt Cancer Center, Tampa, FL; and Mary Ruisi and Joel Jiang, Bristol-Myers Squibb, Princeton, NJ
| | - Bart Neyns
- Sandra P. D'Angelo, Alexander N. Shoushtari, and Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY; James Larkin, Royal Marsden Hospital, London; Paul Lorigan, University of Manchester, Manchester, United Kingdom; Jeffrey A. Sosman, Vanderbilt University Medical Center, Nashville, TN; Celeste Lebbé, Saint-Louis Hospital, Institut National de la Santé et de la Recherche Médicale U976, Université Paris Diderot, Paris; Julie Charles, Grenoble University Hospital, Grenoble Alps University, Grenoble, France; Benjamin Brady, Cabrini Health, Melbourne, Australia; Bart Neyns, Universitair Ziekenhuis Brussel, Brussels, Belgium; Henrik Schmidt, Århus University, Åarhus, Denmark; Jessica C. Hassel, University Hospital Heidelberg, Heidelberg; Peter Mohr, Elbe Kliniken Buxtehude, Buxtehude; Martin Kaatz, SRH Waldklinikum Gera, University Hospital Jena, Jena, Germany; F. Stephen Hodi, Dana-Farber Cancer Institute, Boston, MA; Kerry J. Savage, BC Cancer Agency, University of British Columbia, Vancouver; Wilson H. Miller Jr, Lady Davis Institute and Jewish General Hospital, McGill University, Montreal, Canada; Ivan Marquez-Rodas, Hospital General Universitario Gregorio Marañón, Madrid, Spain; Mario Sznol, Yale University School of Medicine and Smilow Cancer Center, Yale-New Haven Hospital, New Haven, CT; Jeffrey S. Weber, Moffitt Cancer Center, Tampa, FL; and Mary Ruisi and Joel Jiang, Bristol-Myers Squibb, Princeton, NJ
| | - Henrik Schmidt
- Sandra P. D'Angelo, Alexander N. Shoushtari, and Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY; James Larkin, Royal Marsden Hospital, London; Paul Lorigan, University of Manchester, Manchester, United Kingdom; Jeffrey A. Sosman, Vanderbilt University Medical Center, Nashville, TN; Celeste Lebbé, Saint-Louis Hospital, Institut National de la Santé et de la Recherche Médicale U976, Université Paris Diderot, Paris; Julie Charles, Grenoble University Hospital, Grenoble Alps University, Grenoble, France; Benjamin Brady, Cabrini Health, Melbourne, Australia; Bart Neyns, Universitair Ziekenhuis Brussel, Brussels, Belgium; Henrik Schmidt, Århus University, Åarhus, Denmark; Jessica C. Hassel, University Hospital Heidelberg, Heidelberg; Peter Mohr, Elbe Kliniken Buxtehude, Buxtehude; Martin Kaatz, SRH Waldklinikum Gera, University Hospital Jena, Jena, Germany; F. Stephen Hodi, Dana-Farber Cancer Institute, Boston, MA; Kerry J. Savage, BC Cancer Agency, University of British Columbia, Vancouver; Wilson H. Miller Jr, Lady Davis Institute and Jewish General Hospital, McGill University, Montreal, Canada; Ivan Marquez-Rodas, Hospital General Universitario Gregorio Marañón, Madrid, Spain; Mario Sznol, Yale University School of Medicine and Smilow Cancer Center, Yale-New Haven Hospital, New Haven, CT; Jeffrey S. Weber, Moffitt Cancer Center, Tampa, FL; and Mary Ruisi and Joel Jiang, Bristol-Myers Squibb, Princeton, NJ
| | - Jessica C Hassel
- Sandra P. D'Angelo, Alexander N. Shoushtari, and Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY; James Larkin, Royal Marsden Hospital, London; Paul Lorigan, University of Manchester, Manchester, United Kingdom; Jeffrey A. Sosman, Vanderbilt University Medical Center, Nashville, TN; Celeste Lebbé, Saint-Louis Hospital, Institut National de la Santé et de la Recherche Médicale U976, Université Paris Diderot, Paris; Julie Charles, Grenoble University Hospital, Grenoble Alps University, Grenoble, France; Benjamin Brady, Cabrini Health, Melbourne, Australia; Bart Neyns, Universitair Ziekenhuis Brussel, Brussels, Belgium; Henrik Schmidt, Århus University, Åarhus, Denmark; Jessica C. Hassel, University Hospital Heidelberg, Heidelberg; Peter Mohr, Elbe Kliniken Buxtehude, Buxtehude; Martin Kaatz, SRH Waldklinikum Gera, University Hospital Jena, Jena, Germany; F. Stephen Hodi, Dana-Farber Cancer Institute, Boston, MA; Kerry J. Savage, BC Cancer Agency, University of British Columbia, Vancouver; Wilson H. Miller Jr, Lady Davis Institute and Jewish General Hospital, McGill University, Montreal, Canada; Ivan Marquez-Rodas, Hospital General Universitario Gregorio Marañón, Madrid, Spain; Mario Sznol, Yale University School of Medicine and Smilow Cancer Center, Yale-New Haven Hospital, New Haven, CT; Jeffrey S. Weber, Moffitt Cancer Center, Tampa, FL; and Mary Ruisi and Joel Jiang, Bristol-Myers Squibb, Princeton, NJ
| | - F Stephen Hodi
- Sandra P. D'Angelo, Alexander N. Shoushtari, and Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY; James Larkin, Royal Marsden Hospital, London; Paul Lorigan, University of Manchester, Manchester, United Kingdom; Jeffrey A. Sosman, Vanderbilt University Medical Center, Nashville, TN; Celeste Lebbé, Saint-Louis Hospital, Institut National de la Santé et de la Recherche Médicale U976, Université Paris Diderot, Paris; Julie Charles, Grenoble University Hospital, Grenoble Alps University, Grenoble, France; Benjamin Brady, Cabrini Health, Melbourne, Australia; Bart Neyns, Universitair Ziekenhuis Brussel, Brussels, Belgium; Henrik Schmidt, Århus University, Åarhus, Denmark; Jessica C. Hassel, University Hospital Heidelberg, Heidelberg; Peter Mohr, Elbe Kliniken Buxtehude, Buxtehude; Martin Kaatz, SRH Waldklinikum Gera, University Hospital Jena, Jena, Germany; F. Stephen Hodi, Dana-Farber Cancer Institute, Boston, MA; Kerry J. Savage, BC Cancer Agency, University of British Columbia, Vancouver; Wilson H. Miller Jr, Lady Davis Institute and Jewish General Hospital, McGill University, Montreal, Canada; Ivan Marquez-Rodas, Hospital General Universitario Gregorio Marañón, Madrid, Spain; Mario Sznol, Yale University School of Medicine and Smilow Cancer Center, Yale-New Haven Hospital, New Haven, CT; Jeffrey S. Weber, Moffitt Cancer Center, Tampa, FL; and Mary Ruisi and Joel Jiang, Bristol-Myers Squibb, Princeton, NJ
| | - Paul Lorigan
- Sandra P. D'Angelo, Alexander N. Shoushtari, and Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY; James Larkin, Royal Marsden Hospital, London; Paul Lorigan, University of Manchester, Manchester, United Kingdom; Jeffrey A. Sosman, Vanderbilt University Medical Center, Nashville, TN; Celeste Lebbé, Saint-Louis Hospital, Institut National de la Santé et de la Recherche Médicale U976, Université Paris Diderot, Paris; Julie Charles, Grenoble University Hospital, Grenoble Alps University, Grenoble, France; Benjamin Brady, Cabrini Health, Melbourne, Australia; Bart Neyns, Universitair Ziekenhuis Brussel, Brussels, Belgium; Henrik Schmidt, Århus University, Åarhus, Denmark; Jessica C. Hassel, University Hospital Heidelberg, Heidelberg; Peter Mohr, Elbe Kliniken Buxtehude, Buxtehude; Martin Kaatz, SRH Waldklinikum Gera, University Hospital Jena, Jena, Germany; F. Stephen Hodi, Dana-Farber Cancer Institute, Boston, MA; Kerry J. Savage, BC Cancer Agency, University of British Columbia, Vancouver; Wilson H. Miller Jr, Lady Davis Institute and Jewish General Hospital, McGill University, Montreal, Canada; Ivan Marquez-Rodas, Hospital General Universitario Gregorio Marañón, Madrid, Spain; Mario Sznol, Yale University School of Medicine and Smilow Cancer Center, Yale-New Haven Hospital, New Haven, CT; Jeffrey S. Weber, Moffitt Cancer Center, Tampa, FL; and Mary Ruisi and Joel Jiang, Bristol-Myers Squibb, Princeton, NJ
| | - Kerry J Savage
- Sandra P. D'Angelo, Alexander N. Shoushtari, and Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY; James Larkin, Royal Marsden Hospital, London; Paul Lorigan, University of Manchester, Manchester, United Kingdom; Jeffrey A. Sosman, Vanderbilt University Medical Center, Nashville, TN; Celeste Lebbé, Saint-Louis Hospital, Institut National de la Santé et de la Recherche Médicale U976, Université Paris Diderot, Paris; Julie Charles, Grenoble University Hospital, Grenoble Alps University, Grenoble, France; Benjamin Brady, Cabrini Health, Melbourne, Australia; Bart Neyns, Universitair Ziekenhuis Brussel, Brussels, Belgium; Henrik Schmidt, Århus University, Åarhus, Denmark; Jessica C. Hassel, University Hospital Heidelberg, Heidelberg; Peter Mohr, Elbe Kliniken Buxtehude, Buxtehude; Martin Kaatz, SRH Waldklinikum Gera, University Hospital Jena, Jena, Germany; F. Stephen Hodi, Dana-Farber Cancer Institute, Boston, MA; Kerry J. Savage, BC Cancer Agency, University of British Columbia, Vancouver; Wilson H. Miller Jr, Lady Davis Institute and Jewish General Hospital, McGill University, Montreal, Canada; Ivan Marquez-Rodas, Hospital General Universitario Gregorio Marañón, Madrid, Spain; Mario Sznol, Yale University School of Medicine and Smilow Cancer Center, Yale-New Haven Hospital, New Haven, CT; Jeffrey S. Weber, Moffitt Cancer Center, Tampa, FL; and Mary Ruisi and Joel Jiang, Bristol-Myers Squibb, Princeton, NJ
| | - Wilson H Miller
- Sandra P. D'Angelo, Alexander N. Shoushtari, and Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY; James Larkin, Royal Marsden Hospital, London; Paul Lorigan, University of Manchester, Manchester, United Kingdom; Jeffrey A. Sosman, Vanderbilt University Medical Center, Nashville, TN; Celeste Lebbé, Saint-Louis Hospital, Institut National de la Santé et de la Recherche Médicale U976, Université Paris Diderot, Paris; Julie Charles, Grenoble University Hospital, Grenoble Alps University, Grenoble, France; Benjamin Brady, Cabrini Health, Melbourne, Australia; Bart Neyns, Universitair Ziekenhuis Brussel, Brussels, Belgium; Henrik Schmidt, Århus University, Åarhus, Denmark; Jessica C. Hassel, University Hospital Heidelberg, Heidelberg; Peter Mohr, Elbe Kliniken Buxtehude, Buxtehude; Martin Kaatz, SRH Waldklinikum Gera, University Hospital Jena, Jena, Germany; F. Stephen Hodi, Dana-Farber Cancer Institute, Boston, MA; Kerry J. Savage, BC Cancer Agency, University of British Columbia, Vancouver; Wilson H. Miller Jr, Lady Davis Institute and Jewish General Hospital, McGill University, Montreal, Canada; Ivan Marquez-Rodas, Hospital General Universitario Gregorio Marañón, Madrid, Spain; Mario Sznol, Yale University School of Medicine and Smilow Cancer Center, Yale-New Haven Hospital, New Haven, CT; Jeffrey S. Weber, Moffitt Cancer Center, Tampa, FL; and Mary Ruisi and Joel Jiang, Bristol-Myers Squibb, Princeton, NJ
| | - Peter Mohr
- Sandra P. D'Angelo, Alexander N. Shoushtari, and Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY; James Larkin, Royal Marsden Hospital, London; Paul Lorigan, University of Manchester, Manchester, United Kingdom; Jeffrey A. Sosman, Vanderbilt University Medical Center, Nashville, TN; Celeste Lebbé, Saint-Louis Hospital, Institut National de la Santé et de la Recherche Médicale U976, Université Paris Diderot, Paris; Julie Charles, Grenoble University Hospital, Grenoble Alps University, Grenoble, France; Benjamin Brady, Cabrini Health, Melbourne, Australia; Bart Neyns, Universitair Ziekenhuis Brussel, Brussels, Belgium; Henrik Schmidt, Århus University, Åarhus, Denmark; Jessica C. Hassel, University Hospital Heidelberg, Heidelberg; Peter Mohr, Elbe Kliniken Buxtehude, Buxtehude; Martin Kaatz, SRH Waldklinikum Gera, University Hospital Jena, Jena, Germany; F. Stephen Hodi, Dana-Farber Cancer Institute, Boston, MA; Kerry J. Savage, BC Cancer Agency, University of British Columbia, Vancouver; Wilson H. Miller Jr, Lady Davis Institute and Jewish General Hospital, McGill University, Montreal, Canada; Ivan Marquez-Rodas, Hospital General Universitario Gregorio Marañón, Madrid, Spain; Mario Sznol, Yale University School of Medicine and Smilow Cancer Center, Yale-New Haven Hospital, New Haven, CT; Jeffrey S. Weber, Moffitt Cancer Center, Tampa, FL; and Mary Ruisi and Joel Jiang, Bristol-Myers Squibb, Princeton, NJ
| | - Ivan Marquez-Rodas
- Sandra P. D'Angelo, Alexander N. Shoushtari, and Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY; James Larkin, Royal Marsden Hospital, London; Paul Lorigan, University of Manchester, Manchester, United Kingdom; Jeffrey A. Sosman, Vanderbilt University Medical Center, Nashville, TN; Celeste Lebbé, Saint-Louis Hospital, Institut National de la Santé et de la Recherche Médicale U976, Université Paris Diderot, Paris; Julie Charles, Grenoble University Hospital, Grenoble Alps University, Grenoble, France; Benjamin Brady, Cabrini Health, Melbourne, Australia; Bart Neyns, Universitair Ziekenhuis Brussel, Brussels, Belgium; Henrik Schmidt, Århus University, Åarhus, Denmark; Jessica C. Hassel, University Hospital Heidelberg, Heidelberg; Peter Mohr, Elbe Kliniken Buxtehude, Buxtehude; Martin Kaatz, SRH Waldklinikum Gera, University Hospital Jena, Jena, Germany; F. Stephen Hodi, Dana-Farber Cancer Institute, Boston, MA; Kerry J. Savage, BC Cancer Agency, University of British Columbia, Vancouver; Wilson H. Miller Jr, Lady Davis Institute and Jewish General Hospital, McGill University, Montreal, Canada; Ivan Marquez-Rodas, Hospital General Universitario Gregorio Marañón, Madrid, Spain; Mario Sznol, Yale University School of Medicine and Smilow Cancer Center, Yale-New Haven Hospital, New Haven, CT; Jeffrey S. Weber, Moffitt Cancer Center, Tampa, FL; and Mary Ruisi and Joel Jiang, Bristol-Myers Squibb, Princeton, NJ
| | - Julie Charles
- Sandra P. D'Angelo, Alexander N. Shoushtari, and Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY; James Larkin, Royal Marsden Hospital, London; Paul Lorigan, University of Manchester, Manchester, United Kingdom; Jeffrey A. Sosman, Vanderbilt University Medical Center, Nashville, TN; Celeste Lebbé, Saint-Louis Hospital, Institut National de la Santé et de la Recherche Médicale U976, Université Paris Diderot, Paris; Julie Charles, Grenoble University Hospital, Grenoble Alps University, Grenoble, France; Benjamin Brady, Cabrini Health, Melbourne, Australia; Bart Neyns, Universitair Ziekenhuis Brussel, Brussels, Belgium; Henrik Schmidt, Århus University, Åarhus, Denmark; Jessica C. Hassel, University Hospital Heidelberg, Heidelberg; Peter Mohr, Elbe Kliniken Buxtehude, Buxtehude; Martin Kaatz, SRH Waldklinikum Gera, University Hospital Jena, Jena, Germany; F. Stephen Hodi, Dana-Farber Cancer Institute, Boston, MA; Kerry J. Savage, BC Cancer Agency, University of British Columbia, Vancouver; Wilson H. Miller Jr, Lady Davis Institute and Jewish General Hospital, McGill University, Montreal, Canada; Ivan Marquez-Rodas, Hospital General Universitario Gregorio Marañón, Madrid, Spain; Mario Sznol, Yale University School of Medicine and Smilow Cancer Center, Yale-New Haven Hospital, New Haven, CT; Jeffrey S. Weber, Moffitt Cancer Center, Tampa, FL; and Mary Ruisi and Joel Jiang, Bristol-Myers Squibb, Princeton, NJ
| | - Martin Kaatz
- Sandra P. D'Angelo, Alexander N. Shoushtari, and Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY; James Larkin, Royal Marsden Hospital, London; Paul Lorigan, University of Manchester, Manchester, United Kingdom; Jeffrey A. Sosman, Vanderbilt University Medical Center, Nashville, TN; Celeste Lebbé, Saint-Louis Hospital, Institut National de la Santé et de la Recherche Médicale U976, Université Paris Diderot, Paris; Julie Charles, Grenoble University Hospital, Grenoble Alps University, Grenoble, France; Benjamin Brady, Cabrini Health, Melbourne, Australia; Bart Neyns, Universitair Ziekenhuis Brussel, Brussels, Belgium; Henrik Schmidt, Århus University, Åarhus, Denmark; Jessica C. Hassel, University Hospital Heidelberg, Heidelberg; Peter Mohr, Elbe Kliniken Buxtehude, Buxtehude; Martin Kaatz, SRH Waldklinikum Gera, University Hospital Jena, Jena, Germany; F. Stephen Hodi, Dana-Farber Cancer Institute, Boston, MA; Kerry J. Savage, BC Cancer Agency, University of British Columbia, Vancouver; Wilson H. Miller Jr, Lady Davis Institute and Jewish General Hospital, McGill University, Montreal, Canada; Ivan Marquez-Rodas, Hospital General Universitario Gregorio Marañón, Madrid, Spain; Mario Sznol, Yale University School of Medicine and Smilow Cancer Center, Yale-New Haven Hospital, New Haven, CT; Jeffrey S. Weber, Moffitt Cancer Center, Tampa, FL; and Mary Ruisi and Joel Jiang, Bristol-Myers Squibb, Princeton, NJ
| | - Mario Sznol
- Sandra P. D'Angelo, Alexander N. Shoushtari, and Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY; James Larkin, Royal Marsden Hospital, London; Paul Lorigan, University of Manchester, Manchester, United Kingdom; Jeffrey A. Sosman, Vanderbilt University Medical Center, Nashville, TN; Celeste Lebbé, Saint-Louis Hospital, Institut National de la Santé et de la Recherche Médicale U976, Université Paris Diderot, Paris; Julie Charles, Grenoble University Hospital, Grenoble Alps University, Grenoble, France; Benjamin Brady, Cabrini Health, Melbourne, Australia; Bart Neyns, Universitair Ziekenhuis Brussel, Brussels, Belgium; Henrik Schmidt, Århus University, Åarhus, Denmark; Jessica C. Hassel, University Hospital Heidelberg, Heidelberg; Peter Mohr, Elbe Kliniken Buxtehude, Buxtehude; Martin Kaatz, SRH Waldklinikum Gera, University Hospital Jena, Jena, Germany; F. Stephen Hodi, Dana-Farber Cancer Institute, Boston, MA; Kerry J. Savage, BC Cancer Agency, University of British Columbia, Vancouver; Wilson H. Miller Jr, Lady Davis Institute and Jewish General Hospital, McGill University, Montreal, Canada; Ivan Marquez-Rodas, Hospital General Universitario Gregorio Marañón, Madrid, Spain; Mario Sznol, Yale University School of Medicine and Smilow Cancer Center, Yale-New Haven Hospital, New Haven, CT; Jeffrey S. Weber, Moffitt Cancer Center, Tampa, FL; and Mary Ruisi and Joel Jiang, Bristol-Myers Squibb, Princeton, NJ
| | - Jeffrey S Weber
- Sandra P. D'Angelo, Alexander N. Shoushtari, and Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY; James Larkin, Royal Marsden Hospital, London; Paul Lorigan, University of Manchester, Manchester, United Kingdom; Jeffrey A. Sosman, Vanderbilt University Medical Center, Nashville, TN; Celeste Lebbé, Saint-Louis Hospital, Institut National de la Santé et de la Recherche Médicale U976, Université Paris Diderot, Paris; Julie Charles, Grenoble University Hospital, Grenoble Alps University, Grenoble, France; Benjamin Brady, Cabrini Health, Melbourne, Australia; Bart Neyns, Universitair Ziekenhuis Brussel, Brussels, Belgium; Henrik Schmidt, Århus University, Åarhus, Denmark; Jessica C. Hassel, University Hospital Heidelberg, Heidelberg; Peter Mohr, Elbe Kliniken Buxtehude, Buxtehude; Martin Kaatz, SRH Waldklinikum Gera, University Hospital Jena, Jena, Germany; F. Stephen Hodi, Dana-Farber Cancer Institute, Boston, MA; Kerry J. Savage, BC Cancer Agency, University of British Columbia, Vancouver; Wilson H. Miller Jr, Lady Davis Institute and Jewish General Hospital, McGill University, Montreal, Canada; Ivan Marquez-Rodas, Hospital General Universitario Gregorio Marañón, Madrid, Spain; Mario Sznol, Yale University School of Medicine and Smilow Cancer Center, Yale-New Haven Hospital, New Haven, CT; Jeffrey S. Weber, Moffitt Cancer Center, Tampa, FL; and Mary Ruisi and Joel Jiang, Bristol-Myers Squibb, Princeton, NJ
| | - Alexander N Shoushtari
- Sandra P. D'Angelo, Alexander N. Shoushtari, and Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY; James Larkin, Royal Marsden Hospital, London; Paul Lorigan, University of Manchester, Manchester, United Kingdom; Jeffrey A. Sosman, Vanderbilt University Medical Center, Nashville, TN; Celeste Lebbé, Saint-Louis Hospital, Institut National de la Santé et de la Recherche Médicale U976, Université Paris Diderot, Paris; Julie Charles, Grenoble University Hospital, Grenoble Alps University, Grenoble, France; Benjamin Brady, Cabrini Health, Melbourne, Australia; Bart Neyns, Universitair Ziekenhuis Brussel, Brussels, Belgium; Henrik Schmidt, Århus University, Åarhus, Denmark; Jessica C. Hassel, University Hospital Heidelberg, Heidelberg; Peter Mohr, Elbe Kliniken Buxtehude, Buxtehude; Martin Kaatz, SRH Waldklinikum Gera, University Hospital Jena, Jena, Germany; F. Stephen Hodi, Dana-Farber Cancer Institute, Boston, MA; Kerry J. Savage, BC Cancer Agency, University of British Columbia, Vancouver; Wilson H. Miller Jr, Lady Davis Institute and Jewish General Hospital, McGill University, Montreal, Canada; Ivan Marquez-Rodas, Hospital General Universitario Gregorio Marañón, Madrid, Spain; Mario Sznol, Yale University School of Medicine and Smilow Cancer Center, Yale-New Haven Hospital, New Haven, CT; Jeffrey S. Weber, Moffitt Cancer Center, Tampa, FL; and Mary Ruisi and Joel Jiang, Bristol-Myers Squibb, Princeton, NJ
| | - Mary Ruisi
- Sandra P. D'Angelo, Alexander N. Shoushtari, and Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY; James Larkin, Royal Marsden Hospital, London; Paul Lorigan, University of Manchester, Manchester, United Kingdom; Jeffrey A. Sosman, Vanderbilt University Medical Center, Nashville, TN; Celeste Lebbé, Saint-Louis Hospital, Institut National de la Santé et de la Recherche Médicale U976, Université Paris Diderot, Paris; Julie Charles, Grenoble University Hospital, Grenoble Alps University, Grenoble, France; Benjamin Brady, Cabrini Health, Melbourne, Australia; Bart Neyns, Universitair Ziekenhuis Brussel, Brussels, Belgium; Henrik Schmidt, Århus University, Åarhus, Denmark; Jessica C. Hassel, University Hospital Heidelberg, Heidelberg; Peter Mohr, Elbe Kliniken Buxtehude, Buxtehude; Martin Kaatz, SRH Waldklinikum Gera, University Hospital Jena, Jena, Germany; F. Stephen Hodi, Dana-Farber Cancer Institute, Boston, MA; Kerry J. Savage, BC Cancer Agency, University of British Columbia, Vancouver; Wilson H. Miller Jr, Lady Davis Institute and Jewish General Hospital, McGill University, Montreal, Canada; Ivan Marquez-Rodas, Hospital General Universitario Gregorio Marañón, Madrid, Spain; Mario Sznol, Yale University School of Medicine and Smilow Cancer Center, Yale-New Haven Hospital, New Haven, CT; Jeffrey S. Weber, Moffitt Cancer Center, Tampa, FL; and Mary Ruisi and Joel Jiang, Bristol-Myers Squibb, Princeton, NJ
| | - Joel Jiang
- Sandra P. D'Angelo, Alexander N. Shoushtari, and Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY; James Larkin, Royal Marsden Hospital, London; Paul Lorigan, University of Manchester, Manchester, United Kingdom; Jeffrey A. Sosman, Vanderbilt University Medical Center, Nashville, TN; Celeste Lebbé, Saint-Louis Hospital, Institut National de la Santé et de la Recherche Médicale U976, Université Paris Diderot, Paris; Julie Charles, Grenoble University Hospital, Grenoble Alps University, Grenoble, France; Benjamin Brady, Cabrini Health, Melbourne, Australia; Bart Neyns, Universitair Ziekenhuis Brussel, Brussels, Belgium; Henrik Schmidt, Århus University, Åarhus, Denmark; Jessica C. Hassel, University Hospital Heidelberg, Heidelberg; Peter Mohr, Elbe Kliniken Buxtehude, Buxtehude; Martin Kaatz, SRH Waldklinikum Gera, University Hospital Jena, Jena, Germany; F. Stephen Hodi, Dana-Farber Cancer Institute, Boston, MA; Kerry J. Savage, BC Cancer Agency, University of British Columbia, Vancouver; Wilson H. Miller Jr, Lady Davis Institute and Jewish General Hospital, McGill University, Montreal, Canada; Ivan Marquez-Rodas, Hospital General Universitario Gregorio Marañón, Madrid, Spain; Mario Sznol, Yale University School of Medicine and Smilow Cancer Center, Yale-New Haven Hospital, New Haven, CT; Jeffrey S. Weber, Moffitt Cancer Center, Tampa, FL; and Mary Ruisi and Joel Jiang, Bristol-Myers Squibb, Princeton, NJ
| | - Jedd D Wolchok
- Sandra P. D'Angelo, Alexander N. Shoushtari, and Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY; James Larkin, Royal Marsden Hospital, London; Paul Lorigan, University of Manchester, Manchester, United Kingdom; Jeffrey A. Sosman, Vanderbilt University Medical Center, Nashville, TN; Celeste Lebbé, Saint-Louis Hospital, Institut National de la Santé et de la Recherche Médicale U976, Université Paris Diderot, Paris; Julie Charles, Grenoble University Hospital, Grenoble Alps University, Grenoble, France; Benjamin Brady, Cabrini Health, Melbourne, Australia; Bart Neyns, Universitair Ziekenhuis Brussel, Brussels, Belgium; Henrik Schmidt, Århus University, Åarhus, Denmark; Jessica C. Hassel, University Hospital Heidelberg, Heidelberg; Peter Mohr, Elbe Kliniken Buxtehude, Buxtehude; Martin Kaatz, SRH Waldklinikum Gera, University Hospital Jena, Jena, Germany; F. Stephen Hodi, Dana-Farber Cancer Institute, Boston, MA; Kerry J. Savage, BC Cancer Agency, University of British Columbia, Vancouver; Wilson H. Miller Jr, Lady Davis Institute and Jewish General Hospital, McGill University, Montreal, Canada; Ivan Marquez-Rodas, Hospital General Universitario Gregorio Marañón, Madrid, Spain; Mario Sznol, Yale University School of Medicine and Smilow Cancer Center, Yale-New Haven Hospital, New Haven, CT; Jeffrey S. Weber, Moffitt Cancer Center, Tampa, FL; and Mary Ruisi and Joel Jiang, Bristol-Myers Squibb, Princeton, NJ
| |
Collapse
|
5515
|
Ghoneim HE, Zamora AE, Thomas PG, Youngblood BA. Cell-Intrinsic Barriers of T Cell-Based Immunotherapy. Trends Mol Med 2016; 22:1000-1011. [PMID: 27825667 DOI: 10.1016/j.molmed.2016.10.002] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 10/04/2016] [Accepted: 10/05/2016] [Indexed: 12/18/2022]
Abstract
Prolonged exposure of CD8+ T cells to their cognate antigen can result in exhaustion of effector functions enabling the persistence of infected or transformed cells. Recent advances in strategies to rejuvenate host effector function using Immune Checkpoint Blockade have resulted in tremendous success towards the treatment of several cancers. However, it is unclear if T cell rejuvenation results in long-lived antitumor functions. Emerging evidence suggests that T cell exhaustion may also represent a significant impediment in sustaining long-lived antitumor activity by chimeric antigen receptor T cells. Here, we discuss current findings regarding transcriptional regulation during T cell exhaustion and address the hypothesis that epigenetics may be a potential barrier to achieving the maximum benefit of T cell-based immunotherapies.
Collapse
Affiliation(s)
- Hazem E Ghoneim
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA; Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| | - Anthony E Zamora
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Paul G Thomas
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Ben A Youngblood
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA.
| |
Collapse
|
5516
|
Gammaitoni L, Giraudo L, Macagno M, Leuci V, Mesiano G, Rotolo R, Sassi F, Sanlorenzo M, Zaccagna A, Pisacane A, Senetta R, Cangemi M, Cattaneo G, Martin V, Coha V, Gallo S, Pignochino Y, Sapino A, Grignani G, Carnevale-Schianca F, Aglietta M, Sangiolo D. Cytokine-Induced Killer Cells Kill Chemo-surviving Melanoma Cancer Stem Cells. Clin Cancer Res 2016; 23:2277-2288. [PMID: 27815354 DOI: 10.1158/1078-0432.ccr-16-1524] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 10/13/2016] [Accepted: 10/20/2016] [Indexed: 11/16/2022]
Abstract
Purpose: The MHC-unrestricted activity of cytokine-induced killer (CIK) cells against chemo-surviving melanoma cancer stem cells (mCSC) was explored, as CSCs are considered responsible for chemoresistance and relapses.Experimental Design: Putative mCSCs were visualized by engineering patient-derived melanoma cells (MC) with a lentiviral vector encoding eGFP under expression control by stemness gene promoter oct4 Their stemness potential was confirmed in vivo by limiting dilution assays. We explored the sensitivity of eGFP+ mCSCs to chemotherapy (CHT), BRAF inhibitor (BRAFi) or CIK cells, as single agents or in sequence, in vitro First, we treated MCs in vitro with fotemustine or dabrafenib (BRAF-mutated cases); then, surviving MCs, enriched in mCSCs, were challenged with autologous CIK cells. CIK cell activity against chemoresistant mCSCs was confirmed in vivo in two distinct immunodeficient murine models.Results: We visualized eGFP+ mCSCs (14% ± 2.1%) in 11 MCs. The tumorigenic precursor rate in vivo was higher within eGFP+ MCs (1/42) compared with the eGFP- counterpart (1/4,870). In vitro mCSCs were relatively resistant to CHT and BRAFi, but killed by CIK cells (n = 11, 8/11 autologous), with specific lysis ranging from 95% [effector:tumor ratio (E:T), 40:1] to 20% (E:T 1:3). In vivo infusion of autologous CIK cells into mice bearing xenografts from three distinct melanomas demonstrated significant tumor responses involving CHT-spared eGFP+ mCSCs (P = 0.001). Sequential CHT-immunotherapy treatment retained antitumor activity (n = 12, P = 0.001) reducing mCSC rates (P = 0.01).Conclusions: These findings are the first demonstration that immunotherapy with CIK cells is active against autologous mCSCs surviving CHT or BRAFi. An experimental platform for mCSC study and rationale for CIK cells in melanoma clinical study is provided. Clin Cancer Res; 23(9); 2277-88. ©2016 AACR.
Collapse
Affiliation(s)
- Loretta Gammaitoni
- Division of Medical Oncology, Experimental Cell Therapy, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Torino, Italy
| | - Lidia Giraudo
- Division of Medical Oncology, Experimental Cell Therapy, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Torino, Italy
| | - Marco Macagno
- Division of Medical Oncology, Experimental Cell Therapy, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Torino, Italy.,Department of Oncology, University of Torino, Candiolo, Torino, Italy
| | - Valeria Leuci
- Division of Medical Oncology, Experimental Cell Therapy, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Torino, Italy.,Department of Oncology, University of Torino, Candiolo, Torino, Italy
| | - Giulia Mesiano
- Division of Medical Oncology, Experimental Cell Therapy, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Torino, Italy
| | - Ramona Rotolo
- Division of Medical Oncology, Experimental Cell Therapy, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Torino, Italy.,Department of Oncology, University of Torino, Candiolo, Torino, Italy
| | - Francesco Sassi
- Laboratory of Translational Cancer Medicine, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Torino, Italy
| | - Martina Sanlorenzo
- Division of Medical Oncology, Experimental Cell Therapy, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Torino, Italy.,Department of Oncology, University of Torino, Candiolo, Torino, Italy.,Section of Dermatology, Department of Medical Sciences, University of Torino, Torino, Italy
| | - Alessandro Zaccagna
- Surgical Dermatology, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Torino, Italy
| | - Alberto Pisacane
- Pathology, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Torino, Italy
| | - Rebecca Senetta
- Pathology, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Torino, Italy
| | - Michela Cangemi
- Department of Oncology, University of Torino, Candiolo, Torino, Italy
| | - Giulia Cattaneo
- Division of Medical Oncology, Experimental Cell Therapy, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Torino, Italy.,Department of Oncology, University of Torino, Candiolo, Torino, Italy
| | - Valentina Martin
- Division of Medical Oncology, Experimental Cell Therapy, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Torino, Italy
| | - Valentina Coha
- Division of Medical Oncology, Experimental Cell Therapy, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Torino, Italy
| | - Susanna Gallo
- Division of Medical Oncology, Experimental Cell Therapy, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Torino, Italy
| | - Ymera Pignochino
- Division of Medical Oncology, Experimental Cell Therapy, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Torino, Italy.,Laboratory of Translational Cancer Medicine, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Torino, Italy
| | - Anna Sapino
- Pathology, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Torino, Italy
| | - Giovanni Grignani
- Division of Medical Oncology, Experimental Cell Therapy, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Torino, Italy
| | - Fabrizio Carnevale-Schianca
- Division of Medical Oncology, Experimental Cell Therapy, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Torino, Italy
| | - Massimo Aglietta
- Division of Medical Oncology, Experimental Cell Therapy, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Torino, Italy.,Department of Oncology, University of Torino, Candiolo, Torino, Italy
| | - Dario Sangiolo
- Division of Medical Oncology, Experimental Cell Therapy, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Torino, Italy. .,Department of Oncology, University of Torino, Candiolo, Torino, Italy
| |
Collapse
|
5517
|
Johnson DB, Balko JM, Compton ML, Chalkias S, Gorham J, Xu Y, Hicks M, Puzanov I, Alexander MR, Bloomer TL, Becker JR, Slosky DA, Phillips EJ, Pilkinton MA, Craig-Owens L, Kola N, Plautz G, Reshef DS, Deutsch JS, Deering RP, Olenchock BA, Lichtman AH, Roden DM, Seidman CE, Koralnik IJ, Seidman JG, Hoffman RD, Taube JM, Diaz LA, Anders RA, Sosman JA, Moslehi JJ. Fulminant Myocarditis with Combination Immune Checkpoint Blockade. N Engl J Med 2016; 375:1749-1755. [PMID: 27806233 PMCID: PMC5247797 DOI: 10.1056/nejmoa1609214] [Citation(s) in RCA: 1680] [Impact Index Per Article: 186.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Immune checkpoint inhibitors have improved clinical outcomes associated with numerous cancers, but high-grade, immune-related adverse events can occur, particularly with combination immunotherapy. We report the cases of two patients with melanoma in whom fatal myocarditis developed after treatment with ipilimumab and nivolumab. In both patients, there was development of myositis with rhabdomyolysis, early progressive and refractory cardiac electrical instability, and myocarditis with a robust presence of T-cell and macrophage infiltrates. Selective clonal T-cell populations infiltrating the myocardium were identical to those present in tumors and skeletal muscle. Pharmacovigilance studies show that myocarditis occurred in 0.27% of patients treated with a combination of ipilimumab and nivolumab, which suggests that our patients were having a rare, potentially fatal, T-cell-driven drug reaction. (Funded by Vanderbilt-Ingram Cancer Center Ambassadors and others.).
Collapse
Affiliation(s)
- Douglas B Johnson
- From the Departments of Medicine (D.B.J., J.M.B., M.H., I.P., M.R.A., T.L.B., J.R.B., D.A.S., E.J.P., M.A.P., D.M.R., J.A.S., J.J.M.), Cancer Biology (J.M.B., J.J.M.), Pathology, Microbiology, and Immunology (M.L.C., L.C.-O., R.D.H.), Biostatistics (Y.X.), Pharmacology (D.M.R.), and Biomedical Informatics (Y.X., D.M.R.), the Cardio-oncology Program (D.A.S., J.J.M.), the Breast Cancer Research Program (J.M.B.), and the Center for Quantitative Sciences (Y.X.), Vanderbilt University Medical Center, Nashville; the Department of Medicine (S.C.) and the Division of Neuroimmunology (S.C., I.J.K.), Beth Israel Deaconess Medical Center, the Departments of Medicine (B.A.O., C.E.S., J.G.S.) and Pathology (A.H.L.), Brigham and Women's Hospital, the Department of Genetics, Harvard Medical School (J.G., C.E.S., J.G.S.) - all in Boston; Howard Hughes Medical Institute, Chevy Chase, MD (C.E.S.); Bristol-Myers Squibb, New York (N.K., G.P., D.S.R., J.S.D.); Neon Therapeutics, Cambridge, MA (R.P.D.); and the Departments of Pathology (J.M.T., R.A.A.), Dermatology (J.M.T.), and Oncology (L.A.D.), Johns Hopkins University, Baltimore
| | - Justin M Balko
- From the Departments of Medicine (D.B.J., J.M.B., M.H., I.P., M.R.A., T.L.B., J.R.B., D.A.S., E.J.P., M.A.P., D.M.R., J.A.S., J.J.M.), Cancer Biology (J.M.B., J.J.M.), Pathology, Microbiology, and Immunology (M.L.C., L.C.-O., R.D.H.), Biostatistics (Y.X.), Pharmacology (D.M.R.), and Biomedical Informatics (Y.X., D.M.R.), the Cardio-oncology Program (D.A.S., J.J.M.), the Breast Cancer Research Program (J.M.B.), and the Center for Quantitative Sciences (Y.X.), Vanderbilt University Medical Center, Nashville; the Department of Medicine (S.C.) and the Division of Neuroimmunology (S.C., I.J.K.), Beth Israel Deaconess Medical Center, the Departments of Medicine (B.A.O., C.E.S., J.G.S.) and Pathology (A.H.L.), Brigham and Women's Hospital, the Department of Genetics, Harvard Medical School (J.G., C.E.S., J.G.S.) - all in Boston; Howard Hughes Medical Institute, Chevy Chase, MD (C.E.S.); Bristol-Myers Squibb, New York (N.K., G.P., D.S.R., J.S.D.); Neon Therapeutics, Cambridge, MA (R.P.D.); and the Departments of Pathology (J.M.T., R.A.A.), Dermatology (J.M.T.), and Oncology (L.A.D.), Johns Hopkins University, Baltimore
| | - Margaret L Compton
- From the Departments of Medicine (D.B.J., J.M.B., M.H., I.P., M.R.A., T.L.B., J.R.B., D.A.S., E.J.P., M.A.P., D.M.R., J.A.S., J.J.M.), Cancer Biology (J.M.B., J.J.M.), Pathology, Microbiology, and Immunology (M.L.C., L.C.-O., R.D.H.), Biostatistics (Y.X.), Pharmacology (D.M.R.), and Biomedical Informatics (Y.X., D.M.R.), the Cardio-oncology Program (D.A.S., J.J.M.), the Breast Cancer Research Program (J.M.B.), and the Center for Quantitative Sciences (Y.X.), Vanderbilt University Medical Center, Nashville; the Department of Medicine (S.C.) and the Division of Neuroimmunology (S.C., I.J.K.), Beth Israel Deaconess Medical Center, the Departments of Medicine (B.A.O., C.E.S., J.G.S.) and Pathology (A.H.L.), Brigham and Women's Hospital, the Department of Genetics, Harvard Medical School (J.G., C.E.S., J.G.S.) - all in Boston; Howard Hughes Medical Institute, Chevy Chase, MD (C.E.S.); Bristol-Myers Squibb, New York (N.K., G.P., D.S.R., J.S.D.); Neon Therapeutics, Cambridge, MA (R.P.D.); and the Departments of Pathology (J.M.T., R.A.A.), Dermatology (J.M.T.), and Oncology (L.A.D.), Johns Hopkins University, Baltimore
| | - Spyridon Chalkias
- From the Departments of Medicine (D.B.J., J.M.B., M.H., I.P., M.R.A., T.L.B., J.R.B., D.A.S., E.J.P., M.A.P., D.M.R., J.A.S., J.J.M.), Cancer Biology (J.M.B., J.J.M.), Pathology, Microbiology, and Immunology (M.L.C., L.C.-O., R.D.H.), Biostatistics (Y.X.), Pharmacology (D.M.R.), and Biomedical Informatics (Y.X., D.M.R.), the Cardio-oncology Program (D.A.S., J.J.M.), the Breast Cancer Research Program (J.M.B.), and the Center for Quantitative Sciences (Y.X.), Vanderbilt University Medical Center, Nashville; the Department of Medicine (S.C.) and the Division of Neuroimmunology (S.C., I.J.K.), Beth Israel Deaconess Medical Center, the Departments of Medicine (B.A.O., C.E.S., J.G.S.) and Pathology (A.H.L.), Brigham and Women's Hospital, the Department of Genetics, Harvard Medical School (J.G., C.E.S., J.G.S.) - all in Boston; Howard Hughes Medical Institute, Chevy Chase, MD (C.E.S.); Bristol-Myers Squibb, New York (N.K., G.P., D.S.R., J.S.D.); Neon Therapeutics, Cambridge, MA (R.P.D.); and the Departments of Pathology (J.M.T., R.A.A.), Dermatology (J.M.T.), and Oncology (L.A.D.), Johns Hopkins University, Baltimore
| | - Joshua Gorham
- From the Departments of Medicine (D.B.J., J.M.B., M.H., I.P., M.R.A., T.L.B., J.R.B., D.A.S., E.J.P., M.A.P., D.M.R., J.A.S., J.J.M.), Cancer Biology (J.M.B., J.J.M.), Pathology, Microbiology, and Immunology (M.L.C., L.C.-O., R.D.H.), Biostatistics (Y.X.), Pharmacology (D.M.R.), and Biomedical Informatics (Y.X., D.M.R.), the Cardio-oncology Program (D.A.S., J.J.M.), the Breast Cancer Research Program (J.M.B.), and the Center for Quantitative Sciences (Y.X.), Vanderbilt University Medical Center, Nashville; the Department of Medicine (S.C.) and the Division of Neuroimmunology (S.C., I.J.K.), Beth Israel Deaconess Medical Center, the Departments of Medicine (B.A.O., C.E.S., J.G.S.) and Pathology (A.H.L.), Brigham and Women's Hospital, the Department of Genetics, Harvard Medical School (J.G., C.E.S., J.G.S.) - all in Boston; Howard Hughes Medical Institute, Chevy Chase, MD (C.E.S.); Bristol-Myers Squibb, New York (N.K., G.P., D.S.R., J.S.D.); Neon Therapeutics, Cambridge, MA (R.P.D.); and the Departments of Pathology (J.M.T., R.A.A.), Dermatology (J.M.T.), and Oncology (L.A.D.), Johns Hopkins University, Baltimore
| | - Yaomin Xu
- From the Departments of Medicine (D.B.J., J.M.B., M.H., I.P., M.R.A., T.L.B., J.R.B., D.A.S., E.J.P., M.A.P., D.M.R., J.A.S., J.J.M.), Cancer Biology (J.M.B., J.J.M.), Pathology, Microbiology, and Immunology (M.L.C., L.C.-O., R.D.H.), Biostatistics (Y.X.), Pharmacology (D.M.R.), and Biomedical Informatics (Y.X., D.M.R.), the Cardio-oncology Program (D.A.S., J.J.M.), the Breast Cancer Research Program (J.M.B.), and the Center for Quantitative Sciences (Y.X.), Vanderbilt University Medical Center, Nashville; the Department of Medicine (S.C.) and the Division of Neuroimmunology (S.C., I.J.K.), Beth Israel Deaconess Medical Center, the Departments of Medicine (B.A.O., C.E.S., J.G.S.) and Pathology (A.H.L.), Brigham and Women's Hospital, the Department of Genetics, Harvard Medical School (J.G., C.E.S., J.G.S.) - all in Boston; Howard Hughes Medical Institute, Chevy Chase, MD (C.E.S.); Bristol-Myers Squibb, New York (N.K., G.P., D.S.R., J.S.D.); Neon Therapeutics, Cambridge, MA (R.P.D.); and the Departments of Pathology (J.M.T., R.A.A.), Dermatology (J.M.T.), and Oncology (L.A.D.), Johns Hopkins University, Baltimore
| | - Mellissa Hicks
- From the Departments of Medicine (D.B.J., J.M.B., M.H., I.P., M.R.A., T.L.B., J.R.B., D.A.S., E.J.P., M.A.P., D.M.R., J.A.S., J.J.M.), Cancer Biology (J.M.B., J.J.M.), Pathology, Microbiology, and Immunology (M.L.C., L.C.-O., R.D.H.), Biostatistics (Y.X.), Pharmacology (D.M.R.), and Biomedical Informatics (Y.X., D.M.R.), the Cardio-oncology Program (D.A.S., J.J.M.), the Breast Cancer Research Program (J.M.B.), and the Center for Quantitative Sciences (Y.X.), Vanderbilt University Medical Center, Nashville; the Department of Medicine (S.C.) and the Division of Neuroimmunology (S.C., I.J.K.), Beth Israel Deaconess Medical Center, the Departments of Medicine (B.A.O., C.E.S., J.G.S.) and Pathology (A.H.L.), Brigham and Women's Hospital, the Department of Genetics, Harvard Medical School (J.G., C.E.S., J.G.S.) - all in Boston; Howard Hughes Medical Institute, Chevy Chase, MD (C.E.S.); Bristol-Myers Squibb, New York (N.K., G.P., D.S.R., J.S.D.); Neon Therapeutics, Cambridge, MA (R.P.D.); and the Departments of Pathology (J.M.T., R.A.A.), Dermatology (J.M.T.), and Oncology (L.A.D.), Johns Hopkins University, Baltimore
| | - Igor Puzanov
- From the Departments of Medicine (D.B.J., J.M.B., M.H., I.P., M.R.A., T.L.B., J.R.B., D.A.S., E.J.P., M.A.P., D.M.R., J.A.S., J.J.M.), Cancer Biology (J.M.B., J.J.M.), Pathology, Microbiology, and Immunology (M.L.C., L.C.-O., R.D.H.), Biostatistics (Y.X.), Pharmacology (D.M.R.), and Biomedical Informatics (Y.X., D.M.R.), the Cardio-oncology Program (D.A.S., J.J.M.), the Breast Cancer Research Program (J.M.B.), and the Center for Quantitative Sciences (Y.X.), Vanderbilt University Medical Center, Nashville; the Department of Medicine (S.C.) and the Division of Neuroimmunology (S.C., I.J.K.), Beth Israel Deaconess Medical Center, the Departments of Medicine (B.A.O., C.E.S., J.G.S.) and Pathology (A.H.L.), Brigham and Women's Hospital, the Department of Genetics, Harvard Medical School (J.G., C.E.S., J.G.S.) - all in Boston; Howard Hughes Medical Institute, Chevy Chase, MD (C.E.S.); Bristol-Myers Squibb, New York (N.K., G.P., D.S.R., J.S.D.); Neon Therapeutics, Cambridge, MA (R.P.D.); and the Departments of Pathology (J.M.T., R.A.A.), Dermatology (J.M.T.), and Oncology (L.A.D.), Johns Hopkins University, Baltimore
| | - Matthew R Alexander
- From the Departments of Medicine (D.B.J., J.M.B., M.H., I.P., M.R.A., T.L.B., J.R.B., D.A.S., E.J.P., M.A.P., D.M.R., J.A.S., J.J.M.), Cancer Biology (J.M.B., J.J.M.), Pathology, Microbiology, and Immunology (M.L.C., L.C.-O., R.D.H.), Biostatistics (Y.X.), Pharmacology (D.M.R.), and Biomedical Informatics (Y.X., D.M.R.), the Cardio-oncology Program (D.A.S., J.J.M.), the Breast Cancer Research Program (J.M.B.), and the Center for Quantitative Sciences (Y.X.), Vanderbilt University Medical Center, Nashville; the Department of Medicine (S.C.) and the Division of Neuroimmunology (S.C., I.J.K.), Beth Israel Deaconess Medical Center, the Departments of Medicine (B.A.O., C.E.S., J.G.S.) and Pathology (A.H.L.), Brigham and Women's Hospital, the Department of Genetics, Harvard Medical School (J.G., C.E.S., J.G.S.) - all in Boston; Howard Hughes Medical Institute, Chevy Chase, MD (C.E.S.); Bristol-Myers Squibb, New York (N.K., G.P., D.S.R., J.S.D.); Neon Therapeutics, Cambridge, MA (R.P.D.); and the Departments of Pathology (J.M.T., R.A.A.), Dermatology (J.M.T.), and Oncology (L.A.D.), Johns Hopkins University, Baltimore
| | - Tyler L Bloomer
- From the Departments of Medicine (D.B.J., J.M.B., M.H., I.P., M.R.A., T.L.B., J.R.B., D.A.S., E.J.P., M.A.P., D.M.R., J.A.S., J.J.M.), Cancer Biology (J.M.B., J.J.M.), Pathology, Microbiology, and Immunology (M.L.C., L.C.-O., R.D.H.), Biostatistics (Y.X.), Pharmacology (D.M.R.), and Biomedical Informatics (Y.X., D.M.R.), the Cardio-oncology Program (D.A.S., J.J.M.), the Breast Cancer Research Program (J.M.B.), and the Center for Quantitative Sciences (Y.X.), Vanderbilt University Medical Center, Nashville; the Department of Medicine (S.C.) and the Division of Neuroimmunology (S.C., I.J.K.), Beth Israel Deaconess Medical Center, the Departments of Medicine (B.A.O., C.E.S., J.G.S.) and Pathology (A.H.L.), Brigham and Women's Hospital, the Department of Genetics, Harvard Medical School (J.G., C.E.S., J.G.S.) - all in Boston; Howard Hughes Medical Institute, Chevy Chase, MD (C.E.S.); Bristol-Myers Squibb, New York (N.K., G.P., D.S.R., J.S.D.); Neon Therapeutics, Cambridge, MA (R.P.D.); and the Departments of Pathology (J.M.T., R.A.A.), Dermatology (J.M.T.), and Oncology (L.A.D.), Johns Hopkins University, Baltimore
| | - Jason R Becker
- From the Departments of Medicine (D.B.J., J.M.B., M.H., I.P., M.R.A., T.L.B., J.R.B., D.A.S., E.J.P., M.A.P., D.M.R., J.A.S., J.J.M.), Cancer Biology (J.M.B., J.J.M.), Pathology, Microbiology, and Immunology (M.L.C., L.C.-O., R.D.H.), Biostatistics (Y.X.), Pharmacology (D.M.R.), and Biomedical Informatics (Y.X., D.M.R.), the Cardio-oncology Program (D.A.S., J.J.M.), the Breast Cancer Research Program (J.M.B.), and the Center for Quantitative Sciences (Y.X.), Vanderbilt University Medical Center, Nashville; the Department of Medicine (S.C.) and the Division of Neuroimmunology (S.C., I.J.K.), Beth Israel Deaconess Medical Center, the Departments of Medicine (B.A.O., C.E.S., J.G.S.) and Pathology (A.H.L.), Brigham and Women's Hospital, the Department of Genetics, Harvard Medical School (J.G., C.E.S., J.G.S.) - all in Boston; Howard Hughes Medical Institute, Chevy Chase, MD (C.E.S.); Bristol-Myers Squibb, New York (N.K., G.P., D.S.R., J.S.D.); Neon Therapeutics, Cambridge, MA (R.P.D.); and the Departments of Pathology (J.M.T., R.A.A.), Dermatology (J.M.T.), and Oncology (L.A.D.), Johns Hopkins University, Baltimore
| | - David A Slosky
- From the Departments of Medicine (D.B.J., J.M.B., M.H., I.P., M.R.A., T.L.B., J.R.B., D.A.S., E.J.P., M.A.P., D.M.R., J.A.S., J.J.M.), Cancer Biology (J.M.B., J.J.M.), Pathology, Microbiology, and Immunology (M.L.C., L.C.-O., R.D.H.), Biostatistics (Y.X.), Pharmacology (D.M.R.), and Biomedical Informatics (Y.X., D.M.R.), the Cardio-oncology Program (D.A.S., J.J.M.), the Breast Cancer Research Program (J.M.B.), and the Center for Quantitative Sciences (Y.X.), Vanderbilt University Medical Center, Nashville; the Department of Medicine (S.C.) and the Division of Neuroimmunology (S.C., I.J.K.), Beth Israel Deaconess Medical Center, the Departments of Medicine (B.A.O., C.E.S., J.G.S.) and Pathology (A.H.L.), Brigham and Women's Hospital, the Department of Genetics, Harvard Medical School (J.G., C.E.S., J.G.S.) - all in Boston; Howard Hughes Medical Institute, Chevy Chase, MD (C.E.S.); Bristol-Myers Squibb, New York (N.K., G.P., D.S.R., J.S.D.); Neon Therapeutics, Cambridge, MA (R.P.D.); and the Departments of Pathology (J.M.T., R.A.A.), Dermatology (J.M.T.), and Oncology (L.A.D.), Johns Hopkins University, Baltimore
| | - Elizabeth J Phillips
- From the Departments of Medicine (D.B.J., J.M.B., M.H., I.P., M.R.A., T.L.B., J.R.B., D.A.S., E.J.P., M.A.P., D.M.R., J.A.S., J.J.M.), Cancer Biology (J.M.B., J.J.M.), Pathology, Microbiology, and Immunology (M.L.C., L.C.-O., R.D.H.), Biostatistics (Y.X.), Pharmacology (D.M.R.), and Biomedical Informatics (Y.X., D.M.R.), the Cardio-oncology Program (D.A.S., J.J.M.), the Breast Cancer Research Program (J.M.B.), and the Center for Quantitative Sciences (Y.X.), Vanderbilt University Medical Center, Nashville; the Department of Medicine (S.C.) and the Division of Neuroimmunology (S.C., I.J.K.), Beth Israel Deaconess Medical Center, the Departments of Medicine (B.A.O., C.E.S., J.G.S.) and Pathology (A.H.L.), Brigham and Women's Hospital, the Department of Genetics, Harvard Medical School (J.G., C.E.S., J.G.S.) - all in Boston; Howard Hughes Medical Institute, Chevy Chase, MD (C.E.S.); Bristol-Myers Squibb, New York (N.K., G.P., D.S.R., J.S.D.); Neon Therapeutics, Cambridge, MA (R.P.D.); and the Departments of Pathology (J.M.T., R.A.A.), Dermatology (J.M.T.), and Oncology (L.A.D.), Johns Hopkins University, Baltimore
| | - Mark A Pilkinton
- From the Departments of Medicine (D.B.J., J.M.B., M.H., I.P., M.R.A., T.L.B., J.R.B., D.A.S., E.J.P., M.A.P., D.M.R., J.A.S., J.J.M.), Cancer Biology (J.M.B., J.J.M.), Pathology, Microbiology, and Immunology (M.L.C., L.C.-O., R.D.H.), Biostatistics (Y.X.), Pharmacology (D.M.R.), and Biomedical Informatics (Y.X., D.M.R.), the Cardio-oncology Program (D.A.S., J.J.M.), the Breast Cancer Research Program (J.M.B.), and the Center for Quantitative Sciences (Y.X.), Vanderbilt University Medical Center, Nashville; the Department of Medicine (S.C.) and the Division of Neuroimmunology (S.C., I.J.K.), Beth Israel Deaconess Medical Center, the Departments of Medicine (B.A.O., C.E.S., J.G.S.) and Pathology (A.H.L.), Brigham and Women's Hospital, the Department of Genetics, Harvard Medical School (J.G., C.E.S., J.G.S.) - all in Boston; Howard Hughes Medical Institute, Chevy Chase, MD (C.E.S.); Bristol-Myers Squibb, New York (N.K., G.P., D.S.R., J.S.D.); Neon Therapeutics, Cambridge, MA (R.P.D.); and the Departments of Pathology (J.M.T., R.A.A.), Dermatology (J.M.T.), and Oncology (L.A.D.), Johns Hopkins University, Baltimore
| | - Laura Craig-Owens
- From the Departments of Medicine (D.B.J., J.M.B., M.H., I.P., M.R.A., T.L.B., J.R.B., D.A.S., E.J.P., M.A.P., D.M.R., J.A.S., J.J.M.), Cancer Biology (J.M.B., J.J.M.), Pathology, Microbiology, and Immunology (M.L.C., L.C.-O., R.D.H.), Biostatistics (Y.X.), Pharmacology (D.M.R.), and Biomedical Informatics (Y.X., D.M.R.), the Cardio-oncology Program (D.A.S., J.J.M.), the Breast Cancer Research Program (J.M.B.), and the Center for Quantitative Sciences (Y.X.), Vanderbilt University Medical Center, Nashville; the Department of Medicine (S.C.) and the Division of Neuroimmunology (S.C., I.J.K.), Beth Israel Deaconess Medical Center, the Departments of Medicine (B.A.O., C.E.S., J.G.S.) and Pathology (A.H.L.), Brigham and Women's Hospital, the Department of Genetics, Harvard Medical School (J.G., C.E.S., J.G.S.) - all in Boston; Howard Hughes Medical Institute, Chevy Chase, MD (C.E.S.); Bristol-Myers Squibb, New York (N.K., G.P., D.S.R., J.S.D.); Neon Therapeutics, Cambridge, MA (R.P.D.); and the Departments of Pathology (J.M.T., R.A.A.), Dermatology (J.M.T.), and Oncology (L.A.D.), Johns Hopkins University, Baltimore
| | - Nina Kola
- From the Departments of Medicine (D.B.J., J.M.B., M.H., I.P., M.R.A., T.L.B., J.R.B., D.A.S., E.J.P., M.A.P., D.M.R., J.A.S., J.J.M.), Cancer Biology (J.M.B., J.J.M.), Pathology, Microbiology, and Immunology (M.L.C., L.C.-O., R.D.H.), Biostatistics (Y.X.), Pharmacology (D.M.R.), and Biomedical Informatics (Y.X., D.M.R.), the Cardio-oncology Program (D.A.S., J.J.M.), the Breast Cancer Research Program (J.M.B.), and the Center for Quantitative Sciences (Y.X.), Vanderbilt University Medical Center, Nashville; the Department of Medicine (S.C.) and the Division of Neuroimmunology (S.C., I.J.K.), Beth Israel Deaconess Medical Center, the Departments of Medicine (B.A.O., C.E.S., J.G.S.) and Pathology (A.H.L.), Brigham and Women's Hospital, the Department of Genetics, Harvard Medical School (J.G., C.E.S., J.G.S.) - all in Boston; Howard Hughes Medical Institute, Chevy Chase, MD (C.E.S.); Bristol-Myers Squibb, New York (N.K., G.P., D.S.R., J.S.D.); Neon Therapeutics, Cambridge, MA (R.P.D.); and the Departments of Pathology (J.M.T., R.A.A.), Dermatology (J.M.T.), and Oncology (L.A.D.), Johns Hopkins University, Baltimore
| | - Gregory Plautz
- From the Departments of Medicine (D.B.J., J.M.B., M.H., I.P., M.R.A., T.L.B., J.R.B., D.A.S., E.J.P., M.A.P., D.M.R., J.A.S., J.J.M.), Cancer Biology (J.M.B., J.J.M.), Pathology, Microbiology, and Immunology (M.L.C., L.C.-O., R.D.H.), Biostatistics (Y.X.), Pharmacology (D.M.R.), and Biomedical Informatics (Y.X., D.M.R.), the Cardio-oncology Program (D.A.S., J.J.M.), the Breast Cancer Research Program (J.M.B.), and the Center for Quantitative Sciences (Y.X.), Vanderbilt University Medical Center, Nashville; the Department of Medicine (S.C.) and the Division of Neuroimmunology (S.C., I.J.K.), Beth Israel Deaconess Medical Center, the Departments of Medicine (B.A.O., C.E.S., J.G.S.) and Pathology (A.H.L.), Brigham and Women's Hospital, the Department of Genetics, Harvard Medical School (J.G., C.E.S., J.G.S.) - all in Boston; Howard Hughes Medical Institute, Chevy Chase, MD (C.E.S.); Bristol-Myers Squibb, New York (N.K., G.P., D.S.R., J.S.D.); Neon Therapeutics, Cambridge, MA (R.P.D.); and the Departments of Pathology (J.M.T., R.A.A.), Dermatology (J.M.T.), and Oncology (L.A.D.), Johns Hopkins University, Baltimore
| | - Daniel S Reshef
- From the Departments of Medicine (D.B.J., J.M.B., M.H., I.P., M.R.A., T.L.B., J.R.B., D.A.S., E.J.P., M.A.P., D.M.R., J.A.S., J.J.M.), Cancer Biology (J.M.B., J.J.M.), Pathology, Microbiology, and Immunology (M.L.C., L.C.-O., R.D.H.), Biostatistics (Y.X.), Pharmacology (D.M.R.), and Biomedical Informatics (Y.X., D.M.R.), the Cardio-oncology Program (D.A.S., J.J.M.), the Breast Cancer Research Program (J.M.B.), and the Center for Quantitative Sciences (Y.X.), Vanderbilt University Medical Center, Nashville; the Department of Medicine (S.C.) and the Division of Neuroimmunology (S.C., I.J.K.), Beth Israel Deaconess Medical Center, the Departments of Medicine (B.A.O., C.E.S., J.G.S.) and Pathology (A.H.L.), Brigham and Women's Hospital, the Department of Genetics, Harvard Medical School (J.G., C.E.S., J.G.S.) - all in Boston; Howard Hughes Medical Institute, Chevy Chase, MD (C.E.S.); Bristol-Myers Squibb, New York (N.K., G.P., D.S.R., J.S.D.); Neon Therapeutics, Cambridge, MA (R.P.D.); and the Departments of Pathology (J.M.T., R.A.A.), Dermatology (J.M.T.), and Oncology (L.A.D.), Johns Hopkins University, Baltimore
| | - Jonathan S Deutsch
- From the Departments of Medicine (D.B.J., J.M.B., M.H., I.P., M.R.A., T.L.B., J.R.B., D.A.S., E.J.P., M.A.P., D.M.R., J.A.S., J.J.M.), Cancer Biology (J.M.B., J.J.M.), Pathology, Microbiology, and Immunology (M.L.C., L.C.-O., R.D.H.), Biostatistics (Y.X.), Pharmacology (D.M.R.), and Biomedical Informatics (Y.X., D.M.R.), the Cardio-oncology Program (D.A.S., J.J.M.), the Breast Cancer Research Program (J.M.B.), and the Center for Quantitative Sciences (Y.X.), Vanderbilt University Medical Center, Nashville; the Department of Medicine (S.C.) and the Division of Neuroimmunology (S.C., I.J.K.), Beth Israel Deaconess Medical Center, the Departments of Medicine (B.A.O., C.E.S., J.G.S.) and Pathology (A.H.L.), Brigham and Women's Hospital, the Department of Genetics, Harvard Medical School (J.G., C.E.S., J.G.S.) - all in Boston; Howard Hughes Medical Institute, Chevy Chase, MD (C.E.S.); Bristol-Myers Squibb, New York (N.K., G.P., D.S.R., J.S.D.); Neon Therapeutics, Cambridge, MA (R.P.D.); and the Departments of Pathology (J.M.T., R.A.A.), Dermatology (J.M.T.), and Oncology (L.A.D.), Johns Hopkins University, Baltimore
| | - Raquel P Deering
- From the Departments of Medicine (D.B.J., J.M.B., M.H., I.P., M.R.A., T.L.B., J.R.B., D.A.S., E.J.P., M.A.P., D.M.R., J.A.S., J.J.M.), Cancer Biology (J.M.B., J.J.M.), Pathology, Microbiology, and Immunology (M.L.C., L.C.-O., R.D.H.), Biostatistics (Y.X.), Pharmacology (D.M.R.), and Biomedical Informatics (Y.X., D.M.R.), the Cardio-oncology Program (D.A.S., J.J.M.), the Breast Cancer Research Program (J.M.B.), and the Center for Quantitative Sciences (Y.X.), Vanderbilt University Medical Center, Nashville; the Department of Medicine (S.C.) and the Division of Neuroimmunology (S.C., I.J.K.), Beth Israel Deaconess Medical Center, the Departments of Medicine (B.A.O., C.E.S., J.G.S.) and Pathology (A.H.L.), Brigham and Women's Hospital, the Department of Genetics, Harvard Medical School (J.G., C.E.S., J.G.S.) - all in Boston; Howard Hughes Medical Institute, Chevy Chase, MD (C.E.S.); Bristol-Myers Squibb, New York (N.K., G.P., D.S.R., J.S.D.); Neon Therapeutics, Cambridge, MA (R.P.D.); and the Departments of Pathology (J.M.T., R.A.A.), Dermatology (J.M.T.), and Oncology (L.A.D.), Johns Hopkins University, Baltimore
| | - Benjamin A Olenchock
- From the Departments of Medicine (D.B.J., J.M.B., M.H., I.P., M.R.A., T.L.B., J.R.B., D.A.S., E.J.P., M.A.P., D.M.R., J.A.S., J.J.M.), Cancer Biology (J.M.B., J.J.M.), Pathology, Microbiology, and Immunology (M.L.C., L.C.-O., R.D.H.), Biostatistics (Y.X.), Pharmacology (D.M.R.), and Biomedical Informatics (Y.X., D.M.R.), the Cardio-oncology Program (D.A.S., J.J.M.), the Breast Cancer Research Program (J.M.B.), and the Center for Quantitative Sciences (Y.X.), Vanderbilt University Medical Center, Nashville; the Department of Medicine (S.C.) and the Division of Neuroimmunology (S.C., I.J.K.), Beth Israel Deaconess Medical Center, the Departments of Medicine (B.A.O., C.E.S., J.G.S.) and Pathology (A.H.L.), Brigham and Women's Hospital, the Department of Genetics, Harvard Medical School (J.G., C.E.S., J.G.S.) - all in Boston; Howard Hughes Medical Institute, Chevy Chase, MD (C.E.S.); Bristol-Myers Squibb, New York (N.K., G.P., D.S.R., J.S.D.); Neon Therapeutics, Cambridge, MA (R.P.D.); and the Departments of Pathology (J.M.T., R.A.A.), Dermatology (J.M.T.), and Oncology (L.A.D.), Johns Hopkins University, Baltimore
| | - Andrew H Lichtman
- From the Departments of Medicine (D.B.J., J.M.B., M.H., I.P., M.R.A., T.L.B., J.R.B., D.A.S., E.J.P., M.A.P., D.M.R., J.A.S., J.J.M.), Cancer Biology (J.M.B., J.J.M.), Pathology, Microbiology, and Immunology (M.L.C., L.C.-O., R.D.H.), Biostatistics (Y.X.), Pharmacology (D.M.R.), and Biomedical Informatics (Y.X., D.M.R.), the Cardio-oncology Program (D.A.S., J.J.M.), the Breast Cancer Research Program (J.M.B.), and the Center for Quantitative Sciences (Y.X.), Vanderbilt University Medical Center, Nashville; the Department of Medicine (S.C.) and the Division of Neuroimmunology (S.C., I.J.K.), Beth Israel Deaconess Medical Center, the Departments of Medicine (B.A.O., C.E.S., J.G.S.) and Pathology (A.H.L.), Brigham and Women's Hospital, the Department of Genetics, Harvard Medical School (J.G., C.E.S., J.G.S.) - all in Boston; Howard Hughes Medical Institute, Chevy Chase, MD (C.E.S.); Bristol-Myers Squibb, New York (N.K., G.P., D.S.R., J.S.D.); Neon Therapeutics, Cambridge, MA (R.P.D.); and the Departments of Pathology (J.M.T., R.A.A.), Dermatology (J.M.T.), and Oncology (L.A.D.), Johns Hopkins University, Baltimore
| | - Dan M Roden
- From the Departments of Medicine (D.B.J., J.M.B., M.H., I.P., M.R.A., T.L.B., J.R.B., D.A.S., E.J.P., M.A.P., D.M.R., J.A.S., J.J.M.), Cancer Biology (J.M.B., J.J.M.), Pathology, Microbiology, and Immunology (M.L.C., L.C.-O., R.D.H.), Biostatistics (Y.X.), Pharmacology (D.M.R.), and Biomedical Informatics (Y.X., D.M.R.), the Cardio-oncology Program (D.A.S., J.J.M.), the Breast Cancer Research Program (J.M.B.), and the Center for Quantitative Sciences (Y.X.), Vanderbilt University Medical Center, Nashville; the Department of Medicine (S.C.) and the Division of Neuroimmunology (S.C., I.J.K.), Beth Israel Deaconess Medical Center, the Departments of Medicine (B.A.O., C.E.S., J.G.S.) and Pathology (A.H.L.), Brigham and Women's Hospital, the Department of Genetics, Harvard Medical School (J.G., C.E.S., J.G.S.) - all in Boston; Howard Hughes Medical Institute, Chevy Chase, MD (C.E.S.); Bristol-Myers Squibb, New York (N.K., G.P., D.S.R., J.S.D.); Neon Therapeutics, Cambridge, MA (R.P.D.); and the Departments of Pathology (J.M.T., R.A.A.), Dermatology (J.M.T.), and Oncology (L.A.D.), Johns Hopkins University, Baltimore
| | - Christine E Seidman
- From the Departments of Medicine (D.B.J., J.M.B., M.H., I.P., M.R.A., T.L.B., J.R.B., D.A.S., E.J.P., M.A.P., D.M.R., J.A.S., J.J.M.), Cancer Biology (J.M.B., J.J.M.), Pathology, Microbiology, and Immunology (M.L.C., L.C.-O., R.D.H.), Biostatistics (Y.X.), Pharmacology (D.M.R.), and Biomedical Informatics (Y.X., D.M.R.), the Cardio-oncology Program (D.A.S., J.J.M.), the Breast Cancer Research Program (J.M.B.), and the Center for Quantitative Sciences (Y.X.), Vanderbilt University Medical Center, Nashville; the Department of Medicine (S.C.) and the Division of Neuroimmunology (S.C., I.J.K.), Beth Israel Deaconess Medical Center, the Departments of Medicine (B.A.O., C.E.S., J.G.S.) and Pathology (A.H.L.), Brigham and Women's Hospital, the Department of Genetics, Harvard Medical School (J.G., C.E.S., J.G.S.) - all in Boston; Howard Hughes Medical Institute, Chevy Chase, MD (C.E.S.); Bristol-Myers Squibb, New York (N.K., G.P., D.S.R., J.S.D.); Neon Therapeutics, Cambridge, MA (R.P.D.); and the Departments of Pathology (J.M.T., R.A.A.), Dermatology (J.M.T.), and Oncology (L.A.D.), Johns Hopkins University, Baltimore
| | - Igor J Koralnik
- From the Departments of Medicine (D.B.J., J.M.B., M.H., I.P., M.R.A., T.L.B., J.R.B., D.A.S., E.J.P., M.A.P., D.M.R., J.A.S., J.J.M.), Cancer Biology (J.M.B., J.J.M.), Pathology, Microbiology, and Immunology (M.L.C., L.C.-O., R.D.H.), Biostatistics (Y.X.), Pharmacology (D.M.R.), and Biomedical Informatics (Y.X., D.M.R.), the Cardio-oncology Program (D.A.S., J.J.M.), the Breast Cancer Research Program (J.M.B.), and the Center for Quantitative Sciences (Y.X.), Vanderbilt University Medical Center, Nashville; the Department of Medicine (S.C.) and the Division of Neuroimmunology (S.C., I.J.K.), Beth Israel Deaconess Medical Center, the Departments of Medicine (B.A.O., C.E.S., J.G.S.) and Pathology (A.H.L.), Brigham and Women's Hospital, the Department of Genetics, Harvard Medical School (J.G., C.E.S., J.G.S.) - all in Boston; Howard Hughes Medical Institute, Chevy Chase, MD (C.E.S.); Bristol-Myers Squibb, New York (N.K., G.P., D.S.R., J.S.D.); Neon Therapeutics, Cambridge, MA (R.P.D.); and the Departments of Pathology (J.M.T., R.A.A.), Dermatology (J.M.T.), and Oncology (L.A.D.), Johns Hopkins University, Baltimore
| | - Jonathan G Seidman
- From the Departments of Medicine (D.B.J., J.M.B., M.H., I.P., M.R.A., T.L.B., J.R.B., D.A.S., E.J.P., M.A.P., D.M.R., J.A.S., J.J.M.), Cancer Biology (J.M.B., J.J.M.), Pathology, Microbiology, and Immunology (M.L.C., L.C.-O., R.D.H.), Biostatistics (Y.X.), Pharmacology (D.M.R.), and Biomedical Informatics (Y.X., D.M.R.), the Cardio-oncology Program (D.A.S., J.J.M.), the Breast Cancer Research Program (J.M.B.), and the Center for Quantitative Sciences (Y.X.), Vanderbilt University Medical Center, Nashville; the Department of Medicine (S.C.) and the Division of Neuroimmunology (S.C., I.J.K.), Beth Israel Deaconess Medical Center, the Departments of Medicine (B.A.O., C.E.S., J.G.S.) and Pathology (A.H.L.), Brigham and Women's Hospital, the Department of Genetics, Harvard Medical School (J.G., C.E.S., J.G.S.) - all in Boston; Howard Hughes Medical Institute, Chevy Chase, MD (C.E.S.); Bristol-Myers Squibb, New York (N.K., G.P., D.S.R., J.S.D.); Neon Therapeutics, Cambridge, MA (R.P.D.); and the Departments of Pathology (J.M.T., R.A.A.), Dermatology (J.M.T.), and Oncology (L.A.D.), Johns Hopkins University, Baltimore
| | - Robert D Hoffman
- From the Departments of Medicine (D.B.J., J.M.B., M.H., I.P., M.R.A., T.L.B., J.R.B., D.A.S., E.J.P., M.A.P., D.M.R., J.A.S., J.J.M.), Cancer Biology (J.M.B., J.J.M.), Pathology, Microbiology, and Immunology (M.L.C., L.C.-O., R.D.H.), Biostatistics (Y.X.), Pharmacology (D.M.R.), and Biomedical Informatics (Y.X., D.M.R.), the Cardio-oncology Program (D.A.S., J.J.M.), the Breast Cancer Research Program (J.M.B.), and the Center for Quantitative Sciences (Y.X.), Vanderbilt University Medical Center, Nashville; the Department of Medicine (S.C.) and the Division of Neuroimmunology (S.C., I.J.K.), Beth Israel Deaconess Medical Center, the Departments of Medicine (B.A.O., C.E.S., J.G.S.) and Pathology (A.H.L.), Brigham and Women's Hospital, the Department of Genetics, Harvard Medical School (J.G., C.E.S., J.G.S.) - all in Boston; Howard Hughes Medical Institute, Chevy Chase, MD (C.E.S.); Bristol-Myers Squibb, New York (N.K., G.P., D.S.R., J.S.D.); Neon Therapeutics, Cambridge, MA (R.P.D.); and the Departments of Pathology (J.M.T., R.A.A.), Dermatology (J.M.T.), and Oncology (L.A.D.), Johns Hopkins University, Baltimore
| | - Janis M Taube
- From the Departments of Medicine (D.B.J., J.M.B., M.H., I.P., M.R.A., T.L.B., J.R.B., D.A.S., E.J.P., M.A.P., D.M.R., J.A.S., J.J.M.), Cancer Biology (J.M.B., J.J.M.), Pathology, Microbiology, and Immunology (M.L.C., L.C.-O., R.D.H.), Biostatistics (Y.X.), Pharmacology (D.M.R.), and Biomedical Informatics (Y.X., D.M.R.), the Cardio-oncology Program (D.A.S., J.J.M.), the Breast Cancer Research Program (J.M.B.), and the Center for Quantitative Sciences (Y.X.), Vanderbilt University Medical Center, Nashville; the Department of Medicine (S.C.) and the Division of Neuroimmunology (S.C., I.J.K.), Beth Israel Deaconess Medical Center, the Departments of Medicine (B.A.O., C.E.S., J.G.S.) and Pathology (A.H.L.), Brigham and Women's Hospital, the Department of Genetics, Harvard Medical School (J.G., C.E.S., J.G.S.) - all in Boston; Howard Hughes Medical Institute, Chevy Chase, MD (C.E.S.); Bristol-Myers Squibb, New York (N.K., G.P., D.S.R., J.S.D.); Neon Therapeutics, Cambridge, MA (R.P.D.); and the Departments of Pathology (J.M.T., R.A.A.), Dermatology (J.M.T.), and Oncology (L.A.D.), Johns Hopkins University, Baltimore
| | - Luis A Diaz
- From the Departments of Medicine (D.B.J., J.M.B., M.H., I.P., M.R.A., T.L.B., J.R.B., D.A.S., E.J.P., M.A.P., D.M.R., J.A.S., J.J.M.), Cancer Biology (J.M.B., J.J.M.), Pathology, Microbiology, and Immunology (M.L.C., L.C.-O., R.D.H.), Biostatistics (Y.X.), Pharmacology (D.M.R.), and Biomedical Informatics (Y.X., D.M.R.), the Cardio-oncology Program (D.A.S., J.J.M.), the Breast Cancer Research Program (J.M.B.), and the Center for Quantitative Sciences (Y.X.), Vanderbilt University Medical Center, Nashville; the Department of Medicine (S.C.) and the Division of Neuroimmunology (S.C., I.J.K.), Beth Israel Deaconess Medical Center, the Departments of Medicine (B.A.O., C.E.S., J.G.S.) and Pathology (A.H.L.), Brigham and Women's Hospital, the Department of Genetics, Harvard Medical School (J.G., C.E.S., J.G.S.) - all in Boston; Howard Hughes Medical Institute, Chevy Chase, MD (C.E.S.); Bristol-Myers Squibb, New York (N.K., G.P., D.S.R., J.S.D.); Neon Therapeutics, Cambridge, MA (R.P.D.); and the Departments of Pathology (J.M.T., R.A.A.), Dermatology (J.M.T.), and Oncology (L.A.D.), Johns Hopkins University, Baltimore
| | - Robert A Anders
- From the Departments of Medicine (D.B.J., J.M.B., M.H., I.P., M.R.A., T.L.B., J.R.B., D.A.S., E.J.P., M.A.P., D.M.R., J.A.S., J.J.M.), Cancer Biology (J.M.B., J.J.M.), Pathology, Microbiology, and Immunology (M.L.C., L.C.-O., R.D.H.), Biostatistics (Y.X.), Pharmacology (D.M.R.), and Biomedical Informatics (Y.X., D.M.R.), the Cardio-oncology Program (D.A.S., J.J.M.), the Breast Cancer Research Program (J.M.B.), and the Center for Quantitative Sciences (Y.X.), Vanderbilt University Medical Center, Nashville; the Department of Medicine (S.C.) and the Division of Neuroimmunology (S.C., I.J.K.), Beth Israel Deaconess Medical Center, the Departments of Medicine (B.A.O., C.E.S., J.G.S.) and Pathology (A.H.L.), Brigham and Women's Hospital, the Department of Genetics, Harvard Medical School (J.G., C.E.S., J.G.S.) - all in Boston; Howard Hughes Medical Institute, Chevy Chase, MD (C.E.S.); Bristol-Myers Squibb, New York (N.K., G.P., D.S.R., J.S.D.); Neon Therapeutics, Cambridge, MA (R.P.D.); and the Departments of Pathology (J.M.T., R.A.A.), Dermatology (J.M.T.), and Oncology (L.A.D.), Johns Hopkins University, Baltimore
| | - Jeffrey A Sosman
- From the Departments of Medicine (D.B.J., J.M.B., M.H., I.P., M.R.A., T.L.B., J.R.B., D.A.S., E.J.P., M.A.P., D.M.R., J.A.S., J.J.M.), Cancer Biology (J.M.B., J.J.M.), Pathology, Microbiology, and Immunology (M.L.C., L.C.-O., R.D.H.), Biostatistics (Y.X.), Pharmacology (D.M.R.), and Biomedical Informatics (Y.X., D.M.R.), the Cardio-oncology Program (D.A.S., J.J.M.), the Breast Cancer Research Program (J.M.B.), and the Center for Quantitative Sciences (Y.X.), Vanderbilt University Medical Center, Nashville; the Department of Medicine (S.C.) and the Division of Neuroimmunology (S.C., I.J.K.), Beth Israel Deaconess Medical Center, the Departments of Medicine (B.A.O., C.E.S., J.G.S.) and Pathology (A.H.L.), Brigham and Women's Hospital, the Department of Genetics, Harvard Medical School (J.G., C.E.S., J.G.S.) - all in Boston; Howard Hughes Medical Institute, Chevy Chase, MD (C.E.S.); Bristol-Myers Squibb, New York (N.K., G.P., D.S.R., J.S.D.); Neon Therapeutics, Cambridge, MA (R.P.D.); and the Departments of Pathology (J.M.T., R.A.A.), Dermatology (J.M.T.), and Oncology (L.A.D.), Johns Hopkins University, Baltimore
| | - Javid J Moslehi
- From the Departments of Medicine (D.B.J., J.M.B., M.H., I.P., M.R.A., T.L.B., J.R.B., D.A.S., E.J.P., M.A.P., D.M.R., J.A.S., J.J.M.), Cancer Biology (J.M.B., J.J.M.), Pathology, Microbiology, and Immunology (M.L.C., L.C.-O., R.D.H.), Biostatistics (Y.X.), Pharmacology (D.M.R.), and Biomedical Informatics (Y.X., D.M.R.), the Cardio-oncology Program (D.A.S., J.J.M.), the Breast Cancer Research Program (J.M.B.), and the Center for Quantitative Sciences (Y.X.), Vanderbilt University Medical Center, Nashville; the Department of Medicine (S.C.) and the Division of Neuroimmunology (S.C., I.J.K.), Beth Israel Deaconess Medical Center, the Departments of Medicine (B.A.O., C.E.S., J.G.S.) and Pathology (A.H.L.), Brigham and Women's Hospital, the Department of Genetics, Harvard Medical School (J.G., C.E.S., J.G.S.) - all in Boston; Howard Hughes Medical Institute, Chevy Chase, MD (C.E.S.); Bristol-Myers Squibb, New York (N.K., G.P., D.S.R., J.S.D.); Neon Therapeutics, Cambridge, MA (R.P.D.); and the Departments of Pathology (J.M.T., R.A.A.), Dermatology (J.M.T.), and Oncology (L.A.D.), Johns Hopkins University, Baltimore
| |
Collapse
|
5518
|
Johnson DB, Frampton GM, Rioth MJ, Yusko E, Xu Y, Guo X, Ennis RC, Fabrizio D, Chalmers ZR, Greenbowe J, Ali SM, Balasubramanian S, Sun JX, He Y, Frederick DT, Puzanov I, Balko JM, Cates JM, Ross JS, Sanders C, Robins H, Shyr Y, Miller VA, Stephens PJ, Sullivan RJ, Sosman JA, Lovly CM. Targeted Next Generation Sequencing Identifies Markers of Response to PD-1 Blockade. Cancer Immunol Res 2016. [PMID: 27671167 DOI: 10.1158/2326-6066.cir-16-0143.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Therapeutic antibodies blocking programmed death-1 and its ligand (PD-1/PD-L1) induce durable responses in a substantial fraction of melanoma patients. We sought to determine whether the number and/or type of mutations identified using a next-generation sequencing (NGS) panel available in the clinic was correlated with response to anti-PD-1 in melanoma. Using archival melanoma samples from anti-PD-1/PD-L1-treated patients, we performed hybrid capture-based NGS on 236-315 genes and T-cell receptor (TCR) sequencing on initial and validation cohorts from two centers. Patients who responded to anti-PD-1/PD-L1 had higher mutational loads in an initial cohort (median, 45.6 vs. 3.9 mutations/MB; P = 0.003) and a validation cohort (37.1 vs. 12.8 mutations/MB; P = 0.002) compared with nonresponders. Response rate, progression-free survival, and overall survival were superior in the high, compared with intermediate and low, mutation load groups. Melanomas with NF1 mutations harbored high mutational loads (median, 62.7 mutations/MB) and high response rates (74%), whereas BRAF/NRAS/NF1 wild-type melanomas had a lower mutational load. In these archival samples, TCR clonality did not predict response. Mutation numbers in the 315 genes in the NGS platform strongly correlated with those detected by whole-exome sequencing in The Cancer Genome Atlas samples, but was not associated with survival. In conclusion, mutational load, as determined by an NGS platform available in the clinic, effectively stratified patients by likelihood of response. This approach may provide a clinically feasible predictor of response to anti-PD-1/PD-L1. Cancer Immunol Res; 4(11); 959-67. ©2016 AACR.
Collapse
Affiliation(s)
- Douglas B Johnson
- Department of Medicine, Vanderbilt University Medical Center and Vanderbilt Ingram Cancer Center, Nashville, Tennessee.
| | | | - Matthew J Rioth
- Department of Medicine, Vanderbilt University Medical Center and Vanderbilt Ingram Cancer Center, Nashville, Tennessee.,Department of Bioinformatics, Vanderbilt University Medical Center and Vanderbilt Ingram Cancer Center, Nashville, Tennessee
| | - Erik Yusko
- Adaptive Biotechnologies, Seattle, Washington
| | - Yaomin Xu
- Department of Biostatistics, Vanderbilt University Medical Center and Vanderbilt Ingram Cancer Center, Nashville, Tennessee
| | - Xingyi Guo
- Department of Biostatistics, Vanderbilt University Medical Center and Vanderbilt Ingram Cancer Center, Nashville, Tennessee
| | | | | | | | | | - Siraj M Ali
- Foundation Medicine Inc., Cambridge, Massachusetts
| | | | - James X Sun
- Foundation Medicine Inc., Cambridge, Massachusetts
| | - Yuting He
- Foundation Medicine Inc., Cambridge, Massachusetts
| | - Dennie T Frederick
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Igor Puzanov
- Department of Medicine, Vanderbilt University Medical Center and Vanderbilt Ingram Cancer Center, Nashville, Tennessee
| | - Justin M Balko
- Department of Medicine, Vanderbilt University Medical Center and Vanderbilt Ingram Cancer Center, Nashville, Tennessee.,Department of Cancer Biology, Vanderbilt University Medical Center and Vanderbilt Ingram Cancer Center, Nashville, Tennessee
| | - Justin M Cates
- Department of Pathology, Vanderbilt University Medical Center and Vanderbilt Ingram Cancer Center, Nashville, Tennessee
| | | | | | | | - Yu Shyr
- Department of Biostatistics, Vanderbilt University Medical Center and Vanderbilt Ingram Cancer Center, Nashville, Tennessee
| | | | | | - Ryan J Sullivan
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Jeffrey A Sosman
- Department of Medicine, Vanderbilt University Medical Center and Vanderbilt Ingram Cancer Center, Nashville, Tennessee
| | - Christine M Lovly
- Department of Medicine, Vanderbilt University Medical Center and Vanderbilt Ingram Cancer Center, Nashville, Tennessee.,Department of Cancer Biology, Vanderbilt University Medical Center and Vanderbilt Ingram Cancer Center, Nashville, Tennessee
| |
Collapse
|
5519
|
Dillman RO. Is there a role for therapeutic cancer vaccines in the age of checkpoint inhibitors? Hum Vaccin Immunother 2016; 13:528-532. [PMID: 27808593 PMCID: PMC5360141 DOI: 10.1080/21645515.2016.1244149] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Because of the recent success of monoclonal antibody checkpoint inhibitors, and the disappointing results of most therapeutic cancer vaccine trials, it has been questioned whether there is any potential role for such products going forward. In my opinion the answer is “yes” based on the following: [1] there is a persistent unmet clinical need because the majority of patients do not benefit from anti-checkpoint therapy, [2] there is evidence that not all patients make immune responses to their tumors, [3] there is evidence that immune responses to autologous tumor antigens can be induced by patient-specific vaccines, [4] there is clinical evidence from the pre-checkpoint era that suggests survival can be positively impacted by such patient-specific vaccines, and [5] the 2 available therapeutic vaccines that have received regulatory approval are quite limited in terms of their therapeutic benefit.
Collapse
Affiliation(s)
- Robert O Dillman
- a AiVita Biomedical, Inc. , Irvine , CA , USA.,b Hoag Family Cancer Institute , Newport Beach , CA , USA.,c University of California Irvine , Irvine , CA , USA
| |
Collapse
|
5520
|
Shiuan E, Chen J. Eph Receptor Tyrosine Kinases in Tumor Immunity. Cancer Res 2016; 76:6452-6457. [PMID: 27811149 DOI: 10.1158/0008-5472.can-16-1521] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 07/15/2016] [Accepted: 07/20/2016] [Indexed: 12/11/2022]
Abstract
The family of Eph receptor tyrosine kinases and their ephrin ligands regulate a diverse array of physiologic processes, such as axonal guidance, bone remodeling, and immune cell development and trafficking. Eph/ephrin interactions have also been implicated in various pathologic processes, including inflammation, cancer, and tumor angiogenesis. Because Eph receptors play prominent roles in both the immune system and cancer, they likely impact the tumor immune microenvironment, an area in which Eph receptors remain understudied. Here, we provide the first comprehensive review of Eph receptors in the context of tumor immunity. With the recent rise of cancer immunotherapies as promising therapeutic interventions, further elucidation of the roles of Eph receptors in the tumor immune microenvironment will be critical for understanding and developing novel targets against tumor immune evasion. Cancer Res; 76(22); 6452-7. ©2016 AACR.
Collapse
Affiliation(s)
- Eileen Shiuan
- Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, Tennessee.,Medical Scientist Training Program, Vanderbilt University, Nashville, Tennessee
| | - Jin Chen
- Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, Tennessee. .,Division of Rheumatology and Immunology, Department of Medicine, Vanderbilt University, Nashville, Tennessee.,Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee.,Department of Cell & Developmental Biology, Vanderbilt University, Nashville, Tennessee.,Veterans Affairs Medical Center, Tennessee Valley Healthcare System, Nashville, Tennessee
| |
Collapse
|
5521
|
Khalil DN, Postow MA, Ibrahim N, Ludwig DL, Cosaert J, Kambhampati SRP, Tang S, Grebennik D, Kauh JSW, Lenz HJ, Flaherty KT, Hodi FS, Lawrence DP, Wolchok JD. An Open-Label, Dose-Escalation Phase I Study of Anti-TYRP1 Monoclonal Antibody IMC-20D7S for Patients with Relapsed or Refractory Melanoma. Clin Cancer Res 2016; 22:5204-5210. [PMID: 27797971 PMCID: PMC5117650 DOI: 10.1158/1078-0432.ccr-16-1241] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Accepted: 08/04/2016] [Indexed: 11/16/2022]
Abstract
PURPOSE Tyrosinase-related protein-1 (TYRP1) is a transmembrane glycoprotein that is specifically expressed in melanocytes and melanoma cells. Preclinical data suggest that mAbs targeting TYRP1 confer antimelanoma activity. IMC-20D7S is a recombinant human IgG1 mAb targeting TYRP1. Here, we report the first-in-human phase I/Ib trial of IMC-20D7S. EXPERIMENTAL DESIGN The primary objective of this study was to establish the safety profile and the MTD of IMC-20D7S. Patients with advanced melanoma who progressed after or during at least one line of treatment or for whom standard therapy was not indicated enrolled in this standard 3 + 3 dose-escalation, open-label study. IMC-20D7S was administered intravenously every 2 or 3 weeks. RESULTS Twenty-seven patients were enrolled. The most common adverse events were fatigue and constipation experienced by nine (33%) and eight (30%) patients, respectively. There were no serious adverse events related to treatment, no discontinuations of treatment due to adverse events, and no treatment-related deaths. Given the absence of dose-limiting toxicities, an MTD was not defined, but a provisional MTD was established at the 20 mg/kg every 2-week dose based on serum concentration and safety data. One patient experienced a complete response. A disease control rate, defined as stable disease or better, of 41% was observed. CONCLUSION IMC-20D7S is well tolerated among patients with advanced melanoma with evidence of antitumor activity. Further investigation of this agent as monotherapy in selected patients or as part of combination regimens is warranted. Clin Cancer Res; 22(21); 5204-10. ©2016 AACR.
Collapse
Affiliation(s)
- Danny N Khalil
- Memorial Sloan Kettering Cancer Center, Ludwig Center for Cancer Immunotherapy, New York, New York
| | - Michael A Postow
- Memorial Sloan Kettering Cancer Center, Ludwig Center for Cancer Immunotherapy, New York, New York
| | | | | | | | | | | | | | | | - Heinz-Josef Lenz
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California
| | | | - F Stephen Hodi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | | | - Jedd D Wolchok
- Memorial Sloan Kettering Cancer Center, Ludwig Center for Cancer Immunotherapy, New York, New York.
| |
Collapse
|
5522
|
|
5523
|
Schadendorf D, Dummer R, Hauschild A, Robert C, Hamid O, Daud A, van den Eertwegh A, Cranmer L, O'Day S, Puzanov I, Schachter J, Blank C, Salama A, Loquai C, Mehnert JM, Hille D, Ebbinghaus S, Kang SP, Zhou W, Ribas A. Health-related quality of life in the randomised KEYNOTE-002 study of pembrolizumab versus chemotherapy in patients with ipilimumab-refractory melanoma. Eur J Cancer 2016; 67:46-54. [DOI: 10.1016/j.ejca.2016.07.018] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 07/17/2016] [Indexed: 11/28/2022]
|
5524
|
Genetic profiling of hepatocellular carcinoma using next-generation sequencing. J Hepatol 2016; 65:1031-1042. [PMID: 27262756 DOI: 10.1016/j.jhep.2016.05.035] [Citation(s) in RCA: 214] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 04/05/2016] [Accepted: 05/25/2016] [Indexed: 02/06/2023]
Abstract
Hepatocellular carcinoma (HCC) is a highly heterogeneous disease, both clinically and from a molecular standpoint. The advent of next-generation sequencing technologies has provided new opportunities to extensively analyze molecular defects in HCC samples. This has uncovered major cancer driver genes and associated oncogenic pathways operating in HCC. More sophisticated analyses of sequencing data have linked specific nucleotide patterns to external toxic agents and defined so-called 'mutational signatures' in HCC. Molecular signatures, taking into account intra- and inter-tumor heterogeneity, and their functional validation could provide useful data to predict treatment response to molecular therapies. In this review we will focus on the current knowledge of deep sequencing in HCC and its foreseeable clinical impact.
Collapse
|
5525
|
Damuzzo V, Solito S, Pinton L, Carrozzo E, Valpione S, Pigozzo J, Arboretti Giancristofaro R, Chiarion-Sileni V, Mandruzzato S. Clinical implication of tumor-associated and immunological parameters in melanoma patients treated with ipilimumab. Oncoimmunology 2016; 5:e1249559. [PMID: 28123888 PMCID: PMC5215225 DOI: 10.1080/2162402x.2016.1249559] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 09/30/2016] [Accepted: 10/13/2016] [Indexed: 01/05/2023] Open
Abstract
Ipilimumab, the first immune-checkpoint inhibitor extending overall survival (OS) in metastatic melanoma patients, has a survival benefit only in a proportion of patients and the development of reliable predictive biomarkers is still an unmet need. To meet this request, we used a multivariate statistical approach to test whether myeloid-derived suppressor cells (MDSC) or other tumor-associated and immunological parameters may serve as predictive or prognostic biomarkers in melanoma patients receiving ipilimumab. By using a standardized approach to determine the circulating levels of four MDSC subsets, we observed a significant expansion of three MDSC subsets at baseline, as compared to controls and, upon treatment, that high levels of CD14+/IL4Rα+ MDSCs were an independent prognostic factor of reduced OS. On the contrary, longer OS was associated to low levels of the proinflammatory proteins IL-6 and CRP and tumor-associated factors S100B and LDH both at baseline and after treatment. Increasing number of total T cells and especially of PD-1+/CD4+ T cells were associated with better prognosis, and upregulation of PD-1+ expression on CD4+ T cells upon treatment was associated with lower toxicity. As several parameters were associated to OS, we included these factors in a multivariate survival model, and we identified IL-6 and ECOG PS as independent biomarkers associated with improved OS, whereas high levels of LDH and CD14+/IL4Rα+ MDSCs were negative independent markers of reduced OS.
Collapse
Affiliation(s)
- V. Damuzzo
- Oncology and Immunology Section, Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - S. Solito
- Oncology and Immunology Section, Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - L. Pinton
- Oncology and Immunology Section, Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - E. Carrozzo
- Department of Management and Engineering, University of Padova, Padova, Italy
| | - S. Valpione
- Istituto Oncologico Veneto IOV-IRCCS, Padova, Italy
| | - J. Pigozzo
- Istituto Oncologico Veneto IOV-IRCCS, Padova, Italy
| | | | | | - S. Mandruzzato
- Oncology and Immunology Section, Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
- Istituto Oncologico Veneto IOV-IRCCS, Padova, Italy
| |
Collapse
|
5526
|
Lee AY, Droppelmann N, Panageas KS, Zhou Q, Ariyan CE, Brady MS, Chapman PB, Coit DG. Patterns and Timing of Initial Relapse in Pathologic Stage II Melanoma Patients. Ann Surg Oncol 2016; 24:939-946. [PMID: 27804026 DOI: 10.1245/s10434-016-5642-0] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Indexed: 01/26/2023]
Abstract
PURPOSE Pathologic stage II melanoma patients have variable outcomes when divided by substage. We hypothesized that an understanding of the patterns of initial relapse by substage will better inform follow-up guidelines. METHODS We performed a retrospective review of 738 adult patients with pathologic stage II cutaneous melanoma treated at Memorial Sloan Kettering Cancer Center between 1993 and 2013. Clinical records were reviewed to determine time, location, and method of detection of initial relapse. RESULTS At a median follow-up of 52 months, 219 patients relapsed. Relapses were detected more frequently in higher substages. Initial relapses were most commonly local/in-transit for IIA and IIB and systemic for IIC. Lung and brain were the most frequent sites of systemic relapse. Patient-detection was the most common method of relapse detection (59%) in all substages. The 5-year cumulative incidence for patient-detected relapse was 13.6% for IIA, 18.9% for IIB, and 23.3% for IIC and for image-detected relapse was 3.4, 7.9, and 16.6%, respectively. The 5-year cumulative incidence for physician-detected relapse was less than 10% across all substages and leveled off at 3 years for stage IIA and IIB and 2 years for stage IIC. CONCLUSIONS Relapses were most frequently patient-detected in all stage II substages, highlighting the importance of patient education and self-examination. The highest yield for routine imaging is in stage IIC patients during the first 4 years. Physician examination is unlikely to detect relapses beyond 3 years for stage IIA and IIB and beyond 2 years for stage IIC patients.
Collapse
Affiliation(s)
- Ann Y Lee
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,New York University School of Medicine, New York, NY, USA
| | - Nicolas Droppelmann
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Catholic University of Chile School of Medicine, Santiago, Chile
| | - Katherine S Panageas
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Qin Zhou
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Charlotte E Ariyan
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mary S Brady
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Paul B Chapman
- Department of Medical Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Daniel G Coit
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
5527
|
de Filette J, Jansen Y, Schreuer M, Everaert H, Velkeniers B, Neyns B, Bravenboer B. Incidence of Thyroid-Related Adverse Events in Melanoma Patients Treated With Pembrolizumab. J Clin Endocrinol Metab 2016; 101:4431-4439. [PMID: 27571185 PMCID: PMC5095250 DOI: 10.1210/jc.2016-2300] [Citation(s) in RCA: 166] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 08/19/2016] [Indexed: 12/26/2022]
Abstract
CONTEXT Immune checkpoint blockade is associated with endocrine-related adverse events. Thyroid dysfunction during pembrolizumab therapy, an anti-programmed cell death 1 (PD-1) receptor monoclonal antibody, remains to be fully characterized. OBJECTIVE To assess the incidence and characteristics of pembrolizumab-associated thyroid dysfunction. DESIGN AND SETTING Thyroid function was monitored prospectively in melanoma patients who initiated pembrolizumab within an expanded access program at a referral oncology center. 18Fluorodeoxyglucose uptake on positron emission tomography/computed tomography (18FDG-PET/CT) was reviewed in cases compatible with inflammatory thyroiditis. PATIENTS Ninety-nine patients with advanced melanoma (age, 26.3-93.6 years; 63.6% females) who received at least one administration of pembrolizumab. MAIN OUTCOME MEASURES Patient characteristics, thyroid function (TSH, free T4), thyroid autoantibodies, and 18FDG-PET/CT. RESULTS Eighteen adverse events of thyroid dysfunction were observed in 17 patients. Thyrotoxicosis occurred in 12 patients, of which nine evolved to hypothyroidism. Isolated hypothyroidism was present in six patients. Levothyroxine therapy was required in 10 of 15 hypothyroid patients. Thyroid autoantibodies were elevated during thyroid dysfunction in four of 10 cases. Diffuse increased 18FDG uptake by the thyroid gland was observed in all seven thyrotoxic patients who progressed to hypothyroidism. CONCLUSIONS Thyroid dysfunction is common in melanoma patients treated with pembrolizumab. Hypothyroidism and thyrotoxicosis related to inflammatory thyroiditis are the most frequent presentations. Serial measurements of thyroid function tests are indicated during anti-PD-1 monoclonal antibody therapy. Thyrotoxicosis compatible with inflammatory thyroiditis was associated with diffuse increased 18FDG uptake by the thyroid gland. The prospective role of thyroid autoantibodies should be further investigated, together with the histopathological correlates.
Collapse
Affiliation(s)
- Jeroen de Filette
- Departments of Endocrinology (J.d.F., B.V., B.B.), Medical Oncology (Y.J., M.S., B.N.), and Nuclear Medicine (H.E.), Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Yanina Jansen
- Departments of Endocrinology (J.d.F., B.V., B.B.), Medical Oncology (Y.J., M.S., B.N.), and Nuclear Medicine (H.E.), Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Max Schreuer
- Departments of Endocrinology (J.d.F., B.V., B.B.), Medical Oncology (Y.J., M.S., B.N.), and Nuclear Medicine (H.E.), Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Hendrik Everaert
- Departments of Endocrinology (J.d.F., B.V., B.B.), Medical Oncology (Y.J., M.S., B.N.), and Nuclear Medicine (H.E.), Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Brigitte Velkeniers
- Departments of Endocrinology (J.d.F., B.V., B.B.), Medical Oncology (Y.J., M.S., B.N.), and Nuclear Medicine (H.E.), Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Bart Neyns
- Departments of Endocrinology (J.d.F., B.V., B.B.), Medical Oncology (Y.J., M.S., B.N.), and Nuclear Medicine (H.E.), Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Bert Bravenboer
- Departments of Endocrinology (J.d.F., B.V., B.B.), Medical Oncology (Y.J., M.S., B.N.), and Nuclear Medicine (H.E.), Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| |
Collapse
|
5528
|
Brain Metastases From Melanoma: Therapy at the Crossroads. Int J Radiat Oncol Biol Phys 2016; 96:713-716. [DOI: 10.1016/j.ijrobp.2016.06.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 06/01/2016] [Indexed: 12/31/2022]
|
5529
|
Swart M, Verbrugge I, Beltman JB. Combination Approaches with Immune-Checkpoint Blockade in Cancer Therapy. Front Oncol 2016; 6:233. [PMID: 27847783 PMCID: PMC5088186 DOI: 10.3389/fonc.2016.00233] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Accepted: 10/18/2016] [Indexed: 12/11/2022] Open
Abstract
In healthy individuals, immune-checkpoint molecules prevent autoimmune responses and limit immune cell-mediated tissue damage. Tumors frequently exploit these molecules to evade eradication by the immune system. Over the past years, immune-checkpoint blockade of cytotoxic T lymphocyte antigen-4 and programed death-1 emerged as promising strategies to activate antitumor cytotoxic T cell responses. Although complete regression and long-term survival is achieved in some patients, not all patients respond. This review describes promising, novel combination approaches involving immune-checkpoint blockade in the context of the cancer-immunity cycle, aimed at increasing response rates to the single treatments. Specifically, we discuss combinations that promote antigen release and presentation, that further amplify T cell activation, that inhibit trafficking of regulatory T cells or MSDCs, that stimulate intratumoral T cell infiltration, that increase cancer recognition by T cells, and that stimulate tumor killing.
Collapse
Affiliation(s)
- Maarten Swart
- Division of Toxicology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, Netherlands
| | - Inge Verbrugge
- Division of Immunology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Joost B. Beltman
- Division of Toxicology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, Netherlands
| |
Collapse
|
5530
|
Ascierto PA, Vanella V, Grimaldi AM, Lucia F, Palla M, Simeone E, Mozzillo N. Complete response to nivolumab monotherapy in a treatment-naive, BRAF wild-type patient with advanced mucosal melanoma and elevated lactate dehydrogenase: a case report from a phase III trial. Cancer Immunol Immunother 2016; 65:1395-1400. [PMID: 27604993 PMCID: PMC11028877 DOI: 10.1007/s00262-016-1898-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 08/30/2016] [Indexed: 02/03/2023]
Abstract
The anti-PD-1 agent, nivolumab, has been approved both as monotherapy and in combination with ipilimumab for the treatment of unresectable or metastatic melanoma in the USA and European Union. Here we present the case of a patient with treatment-naive, metastatic mucosal melanoma and baseline LDH approximately seven times the upper limit of normal. The patient was enrolled in a clinical trial (CheckMate 066) and achieved a partial response, followed by a durable complete response with nivolumab treatment. The patient's LDH levels were documented in each cycle and dropped markedly within 2 months, when partial response to treatment was already evident. LDH levels remained low for the rest of follow-up, consistent with the ongoing complete response to treatment. The patient experienced only mild immune-related adverse events (grade 1-2), which included vitiligo and rash. This exceptional response suggests that patients with high LDH levels at baseline should be considered for nivolumab treatment. LDH levels, however, should not serve as a predictive marker of response to nivolumab. Moreover, this case suggests the need to identify patients who will achieve the greatest benefit from nivolumab monotherapy.
Collapse
Affiliation(s)
- Paolo A Ascierto
- Istituto Nazionale Tumori Fondazione "G. Pascale", Via Mariano Semmola, 80131, Naples, Italy.
| | - Vito Vanella
- Istituto Nazionale Tumori Fondazione "G. Pascale", Via Mariano Semmola, 80131, Naples, Italy
| | - Antonio Maria Grimaldi
- Istituto Nazionale Tumori Fondazione "G. Pascale", Via Mariano Semmola, 80131, Naples, Italy
| | - Festino Lucia
- Istituto Nazionale Tumori Fondazione "G. Pascale", Via Mariano Semmola, 80131, Naples, Italy
| | - Marco Palla
- Istituto Nazionale Tumori Fondazione "G. Pascale", Via Mariano Semmola, 80131, Naples, Italy
| | - Ester Simeone
- Istituto Nazionale Tumori Fondazione "G. Pascale", Via Mariano Semmola, 80131, Naples, Italy
| | - Nicola Mozzillo
- Istituto Nazionale Tumori Fondazione "G. Pascale", Via Mariano Semmola, 80131, Naples, Italy
| |
Collapse
|
5531
|
Correlation of B7-H3 with androgen receptor, immune pathways and poor outcome in prostate cancer: an expression-based analysis. Prostate Cancer Prostatic Dis 2016; 20:28-35. [PMID: 27801901 DOI: 10.1038/pcan.2016.49] [Citation(s) in RCA: 113] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 08/23/2016] [Accepted: 09/20/2016] [Indexed: 12/19/2022]
Abstract
BACKGROUND B7-H3 (CD276), part of the B7 superfamily of immune checkpoint molecules, has been shown to have an immunomodulatory role. Its regulation, receptor and mechanism of action remain unclear. B7-H3 protein expression correlates with prostate cancer outcomes, and humanized monoclonal antibodies (that is, enoblituzumab) are currently being investigated for therapeutic use. Here we used genomic expression data to examine the relationship between B7-H3 mRNA expression and prostate cancer. METHODS Prostatectomy tissue from 2781 patients were profiled using the Affymetrix HuEx 1.0 ST microarray. Pairwise comparisons were used to identify significant associations between B7-H3 expression and clinicopathologic variables, and survival analyses were used to evaluate the prognostic significance of B7-H3. Pearson's correlation analyses were performed to assess the relationship of B7-H3 expression with molecular subtypes and individual transcripts. Androgen receptor (AR) occupancy at the B7-H3 locus was determined using chromatin immunoprecipitation (ChIP), and androgen-dependent expression changes in B7-H3 was evaluated by quantitative reverse transcription PCR in LNCaP cell lines. Oncomine was queried to evaluate B7-H3 expression in metastatic disease. RESULTS B7-H3 mRNA expression was positively associated with higher Gleason score (P<0.001), tumor stage (P<0.001), and castrate resistant metastatic disease (P<0.0001). High B7-H3 expression correlated with the development of metastasis and prostate cancer specific mortality, but this was not significant on multi-variable analysis. B7-H3 expression correlated with ERG-positive disease (r=0.99) and AR expression (r=0.36). ChIP revealed an AR-binding site upstream of B7-H3, and the presence of androgens decreased B7-H3 expression in LNCaP suggesting potential direct AR regulation. Gene set enrichment analysis demonstrated an association of B7-H3 with androgen signaling as well as immune regulatory pathways. CONCLUSIONS Higher B7-H3 expression correlates with Gleason grade, prostate cancer stage and poor oncologic outcomes in prostatectomy cohorts. B7-H3 expression appears to be related to androgen signaling as well as the immune reactome.
Collapse
|
5532
|
Abstract
PURPOSE OF REVIEW In recent clinical trials, immunotherapeutic agents have demonstrated promising results for the treatment of prostate cancer. This review discusses emerging immunotherapies for prostate cancer and their evolving role in sequencing and combination therapy. RECENT FINDINGS Therapeutic vaccines including PROSTVAC and DCVAC/PCa have completed promising phase 2 trials for the treatment of prostate cancer and phase 3 trials are underway. Recent evidence supports a synergistic relationship between immunotherapy agents themselves, antiandrogens and with cytotoxic chemotherapy. Prostate cancer patients with good prognostic factors, such as minimal disease burden, appear to achieve the optimal benefit from immunotherapy. SUMMARY Therapeutic cancer vaccines and immunomodulating agents have demonstrated activity in the treatment of prostate cancer. Immunotherapies may alter the prostate tumor microenvironment and ongoing studies aim to provide guidance on effective sequencing and combination strategies.
Collapse
|
5533
|
Structural basis of checkpoint blockade by monoclonal antibodies in cancer immunotherapy. Nat Commun 2016; 7:13354. [PMID: 27796306 PMCID: PMC5095608 DOI: 10.1038/ncomms13354] [Citation(s) in RCA: 203] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 09/22/2016] [Indexed: 01/08/2023] Open
Abstract
Cancer cells express tumour-specific antigens derived via genetic and epigenetic alterations, which may be targeted by T-cell-mediated immune responses. However, cancer cells can avoid immune surveillance by suppressing immunity through activation of specific inhibitory signalling pathways, referred to as immune checkpoints. In recent years, the blockade of checkpoint molecules such as PD-1, PD-L1 and CTLA-4, with monoclonal antibodies has enabled the development of breakthrough therapies in oncology, and four therapeutic antibodies targeting these checkpoint molecules have been approved by the FDA for the treatment of several types of cancer. Here, we report the crystal structures of checkpoint molecules in complex with the Fab fragments of therapeutic antibodies, including PD-1/pembrolizumab, PD-1/nivolumab, PD-L1/BMS-936559 and CTLA-4/tremelimumab. These complex structures elucidate the precise epitopes of the antibodies and the molecular mechanisms underlying checkpoint blockade, providing useful information for the improvement of monoclonal antibodies capable of attenuating checkpoint signalling for the treatment of cancer.
Immunotherapy is offering patients with cancer new therapy options. Here, the authors report on the crystal structures of some of these therapies bound to their targets.
Collapse
|
5534
|
Sato Y, Sakurai Y, Kajimoto K, Nakamura T, Yamada Y, Akita H, Harashima H. Innovative Technologies in Nanomedicines: From Passive Targeting to Active Targeting/From Controlled Pharmacokinetics to Controlled Intracellular Pharmacokinetics. Macromol Biosci 2016; 17. [PMID: 27797146 DOI: 10.1002/mabi.201600179] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 07/19/2016] [Indexed: 12/12/2022]
Affiliation(s)
- Yusuke Sato
- Faculty of Pharmaceutical Sciences; Hokkaido University; Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812 Japan
| | - Yu Sakurai
- Faculty of Pharmaceutical Sciences; Hokkaido University; Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812 Japan
| | - Kazuaki Kajimoto
- Health Research Institute; National Institute of Advanced Industrial Science and Technology (AIST); 2217-14 Hayashi-cho Takamatsu, Kagawa 761-0395 Japan
| | - Takashi Nakamura
- Faculty of Pharmaceutical Sciences; Hokkaido University; Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812 Japan
| | - Yuma Yamada
- Faculty of Pharmaceutical Sciences; Hokkaido University; Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812 Japan
| | - Hidetaka Akita
- Graduate School of Pharmaceutical Sciences; Chiba University; 1-8-1 Inohana Chuo-ku, Chiba-shi, Chiba 260-8675 Japan
| | - Hideyoshi Harashima
- Faculty of Pharmaceutical Sciences; Hokkaido University; Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812 Japan
| |
Collapse
|
5535
|
Abstract
Recent advances in genomic sequencing and omics-based capabilities are uncovering tremendous therapeutic opportunities and rapidly transforming the field of cancer medicine. Molecularly targeted agents aim to exploit key tumor-specific vulnerabilities such as oncogenic or non-oncogenic addiction and synthetic lethality. Additionally, immunotherapies targeting the host immune system are proving to be another promising and complementary approach. Owing to substantial tumor genomic and immunologic complexities, combination strategies are likely to be required to adequately disrupt intricate molecular interactions and provide meaningful long-term benefit to patients. To optimize the therapeutic success and application of combination therapies, systematic scientific discovery will need to be coupled with novel and efficient clinical trial approaches. Indeed, a paradigm shift is required to drive precision medicine forward, from the traditional "drug-centric" model of clinical development in pursuit of small incremental benefits in large heterogeneous groups of patients, to a "strategy-centric" model to provide customized transformative treatments in molecularly stratified subsets of patients or even in individual patients. Crucially, to combat the numerous challenges facing combination drug development-including our growing but incomplete understanding of tumor biology, technical and informatics limitations, and escalating financial costs-aligned goals and multidisciplinary collaboration are imperative to collectively harness knowledge and fuel continual innovation.
Collapse
Affiliation(s)
- Daphne Day
- Drug Development Program, Division of Medical Oncology & Hematology, Princess Margaret Cancer Centre, Toronto, Ontario, M5G 2M9, Canada.,Department of Medicine, University of Toronto, Toronto, Ontario, M5S 1A8, Canada.,OICR Research Fellow, Ontario Institute for Cancer Research, Toronto, Ontario, M5G 0A3, Canada
| | - Lillian L Siu
- Drug Development Program, Division of Medical Oncology & Hematology, Princess Margaret Cancer Centre, Toronto, Ontario, M5G 2M9, Canada. .,Department of Medicine, University of Toronto, Toronto, Ontario, M5S 1A8, Canada.
| |
Collapse
|
5536
|
Dronca RS, Mansfield AS, Park SS, Dong H. BCL-2-interacting mediator of cell death (Bim) is a novel biomarker for response to anti-PD-1 therapy in patients with advanced melanoma. Immunotherapy 2016; 8:1351-1353. [PMID: 27784186 DOI: 10.2217/imt-2016-0100] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Affiliation(s)
- Roxana S Dronca
- Division of Medical Oncology, Mayo Clinic, Rochester, MN, USA
| | | | - Sean S Park
- Division of Radiation Oncology, Mayo Clinic, Rochester, MN, USA
| | - Haidong Dong
- Department of Immunology, College of Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA.,Department of Urology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
5537
|
Facteurs prédictifs et biomarqueurs précoces de réponse aux inhibiteurs de checkpoint immunologiques (anti-PD-1, anti-PD-L1). ONCOLOGIE 2016. [DOI: 10.1007/s10269-016-2664-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
5538
|
Grigg C, Blake Z, Gartrell R, Sacher A, Taback B, Saenger Y. Talimogene laherparepvec (T-Vec) for the treatment of melanoma and other cancers. Semin Oncol 2016; 43:638-646. [PMID: 28061981 DOI: 10.1053/j.seminoncol.2016.10.005] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 10/22/2016] [Indexed: 01/14/2023]
Abstract
Talimogene laherparepvec (T-Vec) is the first live virus to be approved by the US Food and Drug Administration for the treatment of cancer. This engineered version of herpes simplex virus type 1 (HSV-1) is the product of decades of preclinical work aimed at identifying and modifying aspects of the viral genome involved in virulence and immunogenicity. T-Vec preferentially infects and lyses tumor cells and, in some cases, induces a systemic immune response against the tumor. These properties have translated into significant and durable clinical responses, particularly in advanced melanoma. Many unanswered questions remain, including how to augment these clinical responses and which other tumor types may respond to oncolytic therapy. Here, we review the development of T-Vec, our current understanding of its impact on the tumor immune micro-environment, and its safety and efficacy in clinical trials for melanoma and other cancers.
Collapse
Affiliation(s)
- Claud Grigg
- Hematology/Oncology, New York-Presbyterian/Columbia University Medical Center, New York, NY
| | - Zoë Blake
- Hematology/Oncology, Columbia University Medical Center, New York, NY
| | - Robyn Gartrell
- Hematology/Oncology, Columbia University Medical Center, New York, NY
| | - Adrian Sacher
- Hematology/Oncology, New York-Presbyterian/Columbia University Medical Center, New York, NY
| | - Bret Taback
- Hematology/Oncology, New York-Presbyterian/Columbia University Medical Center, New York, NY
| | - Yvonne Saenger
- Hematology/Oncology, New York-Presbyterian/Columbia University Medical Center, New York, NY.
| |
Collapse
|
5539
|
Lin J, Wu L, Bai X, Xie Y, Wang A, Zhang H, Yang X, Wan X, Lu X, Sang X, Zhao H. Combination treatment including targeted therapy for advanced hepatocellular carcinoma. Oncotarget 2016; 7:71036-71051. [PMID: 27626176 PMCID: PMC5342607 DOI: 10.18632/oncotarget.11954] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 09/05/2016] [Indexed: 12/14/2022] Open
Abstract
Management of advanced hepatocellular carcinoma (HCC), one of the most lethal cancers worldwide, has presented a therapeutic challenge over past decades. Most patients with advanced HCC and a low possibility of surgical resection have limited treatment options and no alternative but to accept local or palliative treatment. In the new era of cancer therapy, increasing numbers of molecular targeted agents (MTAs) have been applied in the treatment of advanced HCC. However, mono-targeted therapy has shown disappointing outcomes in disease control, primarily because of tumor heterogeneity and complex cell signal transduction. Because incapacitation of a single target is insufficient for cancer suppression, combination treatment for targeted therapy has been proposed and experimentally tested in several clinical trials. In this article, we review research studies aimed to enhance the efficacy of targeted therapy for HCC through combination strategies. Combination treatments involving targeted therapy for advanced HCC are compared and discussed.
Collapse
Affiliation(s)
- Jianzhen Lin
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Liangcai Wu
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Xue Bai
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Yuan Xie
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Anqiang Wang
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Haohai Zhang
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Xiaobo Yang
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Xueshuai Wan
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Xin Lu
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Xinting Sang
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Beijing, China
| | - Haitao Zhao
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), Beijing, China
- Center of Translational Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
5540
|
Eradication of large established tumors in mice by combination immunotherapy that engages innate and adaptive immune responses. Nat Med 2016; 22:1402-1410. [PMID: 27775706 DOI: 10.1038/nm.4200] [Citation(s) in RCA: 421] [Impact Index Per Article: 46.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2016] [Accepted: 09/14/2016] [Indexed: 02/07/2023]
Abstract
Checkpoint blockade with antibodies specific for cytotoxic T lymphocyte-associated protein (CTLA)-4 or programmed cell death 1 (PDCD1; also known as PD-1) elicits durable tumor regression in metastatic cancer, but these dramatic responses are confined to a minority of patients. This suboptimal outcome is probably due in part to the complex network of immunosuppressive pathways present in advanced tumors, which are unlikely to be overcome by intervention at a single signaling checkpoint. Here we describe a combination immunotherapy that recruits a variety of innate and adaptive immune cells to eliminate large tumor burdens in syngeneic tumor models and a genetically engineered mouse model of melanoma; to our knowledge tumors of this size have not previously been curable by treatments relying on endogenous immunity. Maximal antitumor efficacy required four components: a tumor-antigen-targeting antibody, a recombinant interleukin-2 with an extended half-life, anti-PD-1 and a powerful T cell vaccine. Depletion experiments revealed that CD8+ T cells, cross-presenting dendritic cells and several other innate immune cell subsets were required for tumor regression. Effective treatment induced infiltration of immune cells and production of inflammatory cytokines in the tumor, enhanced antibody-mediated tumor antigen uptake and promoted antigen spreading. These results demonstrate the capacity of an elicited endogenous immune response to destroy large, established tumors and elucidate essential characteristics of combination immunotherapies that are capable of curing a majority of tumors in experimental settings typically viewed as intractable.
Collapse
|
5541
|
Pastor F. Aptamers: A New Technological Platform in Cancer Immunotherapy. Pharmaceuticals (Basel) 2016; 9:E64. [PMID: 27783034 PMCID: PMC5198039 DOI: 10.3390/ph9040064] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Revised: 09/29/2016] [Accepted: 10/19/2016] [Indexed: 12/12/2022] Open
Abstract
The renaissance of cancer immunotherapy is, nowadays, a reality. In the near future, it will be very likely among the first-line treatments for cancer patients. There are several different approaches to modulate the immune system to fight against tumor maladies but, so far, monoclonal antibodies may currently be the most successful immuno-tools used to that end. The number of ongoing clinical trials with monoclonal antibodies has been increasing exponentially over the last few years upon the Food and Drug Administration (FDA) approval of the first immune-checkpoint blockade antibodies. In spite of the proved antitumor effect of these reagents, the unleashing of the immune system to fight cancer cells has a cost, namely auto-inflammatory toxicity. Additionally, only a small fraction of all patients treated with immune-checkpoint antibodies have a clinical benefit. Taking into account all this, it is urgent new therapeutic reagents are developed with a contained toxicity that could facilitate the combination of different immune-modulating pathways to broaden the antitumor effect in most cancer patients. Based on preclinical data, oligonucleotide aptamers could fulfill this need. Aptamers have not only been successfully used as antagonists of immune-checkpoint receptors, but also as agonists of immunostimulatory receptors in cancer immunotherapy. The simplicity of aptamers to be engineered for the specific delivery of different types of cargos to tumor cells and immune cells so as to harvest an efficient antitumor immune response gives aptamers a significant advantage over antibodies. In this review all of the recent applications of aptamers in cancer immunotherapy will be described.
Collapse
Affiliation(s)
- Fernando Pastor
- Instituto de Investigación Sanitaria de Navarra (IDISNA), Recinto de Complejo Hospitalario de Navarra, Pamplona 31008, Spain.
- Program of Molecular Therapies, Aptamer Unit, Centro de Investigación Medica Aplicada (CIMA), Pamplona 31008, Spain.
| |
Collapse
|
5542
|
Kyte JA, Gaudernack G, Faane A, Lislerud K, Inderberg EM, Brunsvig P, Aamdal S, Kvalheim G, Wälchli S, Pule M. T-helper cell receptors from long-term survivors after telomerase cancer vaccination for use in adoptive cell therapy. Oncoimmunology 2016; 5:e1249090. [PMID: 28123886 DOI: 10.1080/2162402x.2016.1249090] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 09/28/2016] [Accepted: 10/12/2016] [Indexed: 12/17/2022] Open
Abstract
We herein report retargeting of T-helper (Th) cells against the universal cancer antigen telomerase for use in adoptive cell therapy. The redirected Th cells may counter tumor tolerance, transform the inflammatory milieu, and induce epitope spreading and cancer senescence. We have previously conducted a series of trials evaluating vaccination with telomerase peptides. From long-term survivors, we isolated >100 CD4+ Th-cell clones recognizing telomerase epitopes. The clones were characterized with regard to HLA restriction, functional avidity, fine specificity, proliferative capacity, cytokine profile, and recognition of naturally processed epitopes. DP4 is the most prevalent HLA molecule worldwide. Two DP4-restricted T-cell clones with different functional avidity, C13 and D71, were selected for molecular T-cell receptor (TCR) cloning. Both clones showed a high proliferative capacity, recognition of naturally processed telomerase epitopes, and a polyfunctional and Th1-weighted cytokine profile. TCR C13 and D71 were cloned into the retroviral vector MP71 together with the compact and GMP-applicable marker/suicide gene RQR8. Both TCRs were expressed well in recipient T cells after PBMC transduction. The transduced T cells co-expressed RQR8 and acquired the desired telomerase specificity, with a polyfunctional response including production of TNFa, IFNγ, and CD107a. Interestingly, the DP4-restricted TCRs were expressed and functional both in CD4+ and CD8+ T cells. The findings demonstrate that the cloned TCRs confer recipient T cells with the desired hTERT-specificity and functionality. We hypothesize that adoptive therapy with Th cells may offer a powerful novel approach for overcoming tumor tolerance and synergize with other forms of immunotherapy.
Collapse
Affiliation(s)
- Jon Amund Kyte
- Department for Cell Therapy, Oslo University Hospital, Oslo, Norway; Department of Oncology, Oslo University Hospital, Oslo, Norway; Department of Haematology, UCL Cancer Institute, University College London, London, UK
| | - Gustav Gaudernack
- Department for Immunology, Cancer Research Institute, Oslo University Hospital, Oslo, Norway; Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Anne Faane
- Department for Cell Therapy, Oslo University Hospital , Oslo, Norway
| | - Kari Lislerud
- Department for Cell Therapy, Oslo University Hospital , Oslo, Norway
| | | | - Paal Brunsvig
- Clinical Trial Unit, Department of Oncology, Oslo University Hospital , Oslo, Norway
| | - Steinar Aamdal
- Faculty of Medicine, University of Oslo, Oslo, Norway; Clinical Trial Unit, Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Gunnar Kvalheim
- Department for Cell Therapy, Oslo University Hospital , Oslo, Norway
| | - Sébastien Wälchli
- Department for Cell Therapy, Oslo University Hospital, Oslo, Norway; Department for Immunology, Cancer Research Institute, Oslo University Hospital, Oslo, Norway
| | - Martin Pule
- Department of Haematology, UCL Cancer Institute, University College London , London, UK
| |
Collapse
|
5543
|
Ramelyte E, Schindler SA, Dummer R. The safety of anti PD-1 therapeutics for the treatment of melanoma. Expert Opin Drug Saf 2016; 16:41-53. [PMID: 27737598 DOI: 10.1080/14740338.2016.1248402] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
INTRODUCTION The introduction of immunotherapies into clinical practice has substantially improved the prognosis of metastatic melanoma patients as well as patients suffering from other cancers. The two FDA-approved checkpoint inhibitors against PD-1 (nivolumab and pembrolizumab) have been shown to significantly improve patient survival while being less toxic than previous treatment options. Areas covered: The current scientific literature on safety and adverse events (AEs) related to anti-PD-1 therapies has been investigated with special attention to case reports and to the latest results announced at the major clinical cancer and melanoma meetings, including ASCO (American Society of Clinical Oncology), ESMO (European Society of medical Oncology) and EADO (European Association of Dermato-Oncology) annual meetings. Expert opinion: Even though anti-PD-1 therapies are better tolerated than conventional chemo- or other immune-therapies, they still induce a plethora of AEs. Given the mechanism of action, it is supposed that most if not all of them are immune related. Fortunately, the majority are mild and manageable. However, due to the increase in patients' life expectancy, there is a substantial need to understand and prevent severe cutaneous, pulmonary, neurological and other AEs which have major impact on the quality of life. The safety profile after long term use of these medications is still unclear. In addition, non-steroid based immune interventions to control autoimmunity are still to be developed.
Collapse
Affiliation(s)
- Egle Ramelyte
- a Department of Dermatology , University Hospital Zurich , Zurich , Switzerland.,b Centre of Dermatovenereology , Vilnius University Hospital Santariskiu klinikos , Vilnius , Lithuania
| | - Sabrina A Schindler
- a Department of Dermatology , University Hospital Zurich , Zurich , Switzerland
| | - Reinhard Dummer
- a Department of Dermatology , University Hospital Zurich , Zurich , Switzerland
| |
Collapse
|
5544
|
Cohen JV, Tawbi H, Margolin KA, Amravadi R, Bosenberg M, Brastianos PK, Chiang VL, de Groot J, Glitza IC, Herlyn M, Holmen SL, Jilaveanu LB, Lassman A, Moschos S, Postow MA, Thomas R, Tsiouris JA, Wen P, White RM, Turnham T, Davies MA, Kluger HM. Melanoma central nervous system metastases: current approaches, challenges, and opportunities. Pigment Cell Melanoma Res 2016; 29:627-642. [PMID: 27615400 DOI: 10.1111/pcmr.12538] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 09/06/2016] [Indexed: 12/17/2022]
Abstract
Melanoma central nervous system metastases are increasing, and the challenges presented by this patient population remain complex. In December 2015, the Melanoma Research Foundation and the Wistar Institute hosted the First Summit on Melanoma Central Nervous System (CNS) Metastases in Philadelphia, Pennsylvania. Here, we provide a review of the current status of the field of melanoma brain metastasis research; identify key challenges and opportunities for improving the outcomes in patients with melanoma brain metastases; and set a framework to optimize future research in this critical area.
Collapse
Affiliation(s)
- Justine V Cohen
- Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA
| | - Hussain Tawbi
- Department of Melanoma, Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kim A Margolin
- Department of Medical Oncology & Therapeutics Research, City of Hope Cancer Center, Duarte, CA, USA
| | - Ravi Amravadi
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | | | | | | | - John de Groot
- Division of Neuro-Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - Isabella C Glitza
- Department of Melanoma, Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Meenhard Herlyn
- Melanoma Research Center, The Wistar Institute, Philadelphia, PA, USA
| | - Sheri L Holmen
- Huntsman Cancer Institute, University of Utah Health Sciences Center, Salt Lake City, UT, USA
| | | | - Andrew Lassman
- Department of Neurology & Herbert Irving Comprehensive, Cancer Center, Columbia University Medical Center, New York, NY, USA
| | - Stergios Moschos
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Michael A Postow
- Department of Oncology, Memorial Sloan Kettering Cancer Center and Weill Cornell Medicine, New York, NY, USA
| | - Reena Thomas
- Division of Neuro-Oncology, Department of Neurology, Stanford University, Stanford, CA, USA
| | - John A Tsiouris
- Department of Radiology, New York-Presbyterian Hospital - Weill Cornell Medicine, New York, NY, USA
| | - Patrick Wen
- Department of Neuro-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Richard M White
- Department of Cancer Biology & Genetics, Memorial Sloan Kettering Cancer Center and Weill Cornell Medicine, New York, NY, USA
| | | | - Michael A Davies
- Department of Melanoma, Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | |
Collapse
|
5545
|
Nivolumab for melanoma, non-small cell lung cancer. Aust Prescr 2016; 39:138-140. [PMID: 27756979 DOI: 10.18773/austprescr.2016.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
|
5546
|
Dagogo-Jack I, Gill CM, Cahill DP, Santagata S, Brastianos PK. Treatment of brain metastases in the modern genomic era. Pharmacol Ther 2016; 170:64-72. [PMID: 27773784 DOI: 10.1016/j.pharmthera.2016.10.011] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Development of brain metastasis (BM) portends a dismal prognosis for patients with cancer. Melanomas and carcinomas of the lung, breast, and kidney are the most common malignancies to metastasize to the brain. Recent advances in molecular genetics have enabled the identification of actionable, clinically relevant genetic alterations within primary tumors and their corresponding metastases. Adoption of genotype-guided treatment strategies for the management of systemic malignancy has resulted in dramatic and durable responses. Unfortunately, despite these therapeutic advances, central nervous system (CNS) relapses are not uncommon. Although these relapses have historically been attributed to limited blood brain barrier penetration of anti-neoplastic agents, recent work has demonstrated genetic heterogeneity such that metastatic sites, including BM, harbor relevant genetic alterations that are not present in primary tumor biopsies. This improved insight into molecular mechanisms underlying site specific recurrences can inform strategies for targeting these oncogenic drivers. Thus, development of rational, genomically guided CNS-penetrant therapies is crucial for ongoing therapeutic success.
Collapse
Affiliation(s)
- Ibiayi Dagogo-Jack
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States; Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Corey M Gill
- Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States; Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Daniel P Cahill
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Sandro Santagata
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Priscilla K Brastianos
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States; Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States; Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
5547
|
Temel JS, Shaw AT, Greer JA. Challenge of Prognostic Uncertainty in the Modern Era of Cancer Therapeutics. J Clin Oncol 2016; 34:3605-3608. [PMID: 27551112 DOI: 10.1200/jco.2016.67.8573] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
5548
|
Kyte JA, Aamdal S, Dueland S, Sæbøe-Larsen S, Inderberg EM, Madsbu UE, Skovlund E, Gaudernack G, Kvalheim G. Immune response and long-term clinical outcome in advanced melanoma patients vaccinated with tumor-mRNA-transfected dendritic cells. Oncoimmunology 2016; 5:e1232237. [PMID: 27999747 DOI: 10.1080/2162402x.2016.1232237] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 08/30/2016] [Accepted: 08/31/2016] [Indexed: 01/18/2023] Open
Abstract
The most effective anticancer immune responses are probably directed against patient-specific neoantigens. We have developed a melanoma vaccine targeting this individual mutanome based on dendritic cells (DCs) loaded with autologous tumor-mRNA. Here, we report a phase I/II trial evaluating toxicity, immune response and clinical outcome in 31 metastatic melanoma patients. The first cohort (n = 22) received the vaccine without any adjuvant; the next cohort (n = 9) received adjuvant IL2. Each subject received four weekly intranodal or intradermal injections, followed by optional monthly vaccines. Immune response was evaluated by delayed-type hypersensitivity (DTH), T cell proliferation and cytokine assays. Data were collected for 10 y after inclusion of the last patient. No serious adverse events were detected. In the intention-to-treat-cohort, we demonstrated significantly superior survival compared to matched controls from a benchmark meta-analysis (1 y survival 43% vs. 24%, 2 y 23% vs. 6.6%). A tumor-specific immune response was demonstrated in 16/31 patients. The response rate was higher after intradermal than intranodal vaccination (80% vs. 38%). Immune responders had improved survival compared to non-responders (median 14 mo vs. 6 mo; p = 0.030), and all eight patients surviving >20 mo were immune responders. In addition to the tumor-specific response, most patients developed a response against autologous DC antigens. The cytokine profile was polyfunctional and did not follow a Th1/Th2 dichotomy. We conclude that the favorable safety profile and evidence of a possible survival benefit warrant further studies of the RNA/DC vaccine. The vaccine appears insufficient as monotherapy, but there is a strong rationale for combination with checkpoint modulators.
Collapse
Affiliation(s)
- Jon Amund Kyte
- Department for Cell Therapy, Radiumhospitalet, Oslo University Hospital, Oslo, Norway; The Clinical Trial Unit, Radiumhospitalet, Oslo University Hospital, Oslo, Norway; Department of Immunology, Radiumhospitalet, Oslo University Hospital, Oslo, Norway
| | - Steinar Aamdal
- The Clinical Trial Unit, Radiumhospitalet, Oslo University Hospital , Oslo, Norway
| | - Svein Dueland
- The Clinical Trial Unit, Radiumhospitalet, Oslo University Hospital , Oslo, Norway
| | - Stein Sæbøe-Larsen
- Department for Cell Therapy, Radiumhospitalet, Oslo University Hospital , Oslo, Norway
| | - Else Marit Inderberg
- Department for Cell Therapy, Radiumhospitalet, Oslo University Hospital , Oslo, Norway
| | - Ulf Erik Madsbu
- Department for Radiology, Radiumhospitalet, Oslo University Hospital , Oslo, Norway
| | - Eva Skovlund
- Department of Public Health and General Practice, NTNU , Trondheim, Norway
| | - Gustav Gaudernack
- Department of Immunology, Radiumhospitalet, Oslo University Hospital , Oslo, Norway
| | - Gunnar Kvalheim
- Department for Cell Therapy, Radiumhospitalet, Oslo University Hospital , Oslo, Norway
| |
Collapse
|
5549
|
Johnpulle RAN, Conry RM, Sosman JA, Puzanov I, Johnson DB. Responses to immune checkpoint inhibitors in nonagenarians. Oncoimmunology 2016; 5:e1234572. [PMID: 27999751 PMCID: PMC5139628 DOI: 10.1080/2162402x.2016.1234572] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 08/29/2016] [Accepted: 09/05/2016] [Indexed: 10/20/2022] Open
Abstract
The incidence of melanoma continues to rise with the most rapid increase seen in the elderly population. Historically, elderly patients with advanced melanoma have had dismal clinical outcomes, in part, due to distinct tumor biology, and often ineligibility for effective therapies during their development. In addition, due to relatively few geriatric patients being accrued to clinical trials of novel immunotherapeutics, there is a paucity of data regarding their safety and efficacy. Herein, we present the clinical course of three consecutive nonagenarians (≥90 y old) with metastatic melanoma, who were treated with single-agent or combination immune checkpoint inhibitors. Two patients experienced complete or partial responses with acceptable safety profiles, and one other tolerated therapy well although a significant response was not noted. These cases suggest that with close monitoring, even very elderly patients with advanced cancers and acceptable performance status may tolerate and benefit from immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Romany A. N. Johnpulle
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA,CONTACT Romany A. N. Johnpulle Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Robert M. Conry
- The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jeffrey A. Sosman
- Robert H Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA
| | | | - Douglas B. Johnson
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
5550
|
Furudate S, Fujimura T, Kambayashi Y, Hidaka T, Hashimoto A, Aiba S. Sequential Therapy with Nivolumab Followed by Ipilimumab Induces Complete Response in Metastatic Melanoma of the Lung but with Severe Hepatotoxicities. Case Rep Oncol 2016; 9:644-649. [PMID: 27920696 PMCID: PMC5118831 DOI: 10.1159/000450974] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Accepted: 09/21/2016] [Indexed: 11/22/2022] Open
Abstract
Since nivolumab significantly prolongs survival in patients with metastatic melanoma, the number of patients administered nivolumab is increasing, but only 30–40% of patients who received nivolumab monotherapy experienced objective tumor regression. Therefore, enhancing its anti-tumor immune response is of great interest to dermato-oncologists. In this report, we present a case of multiple metastatic melanomas in the lung successfully treated with nivolumab (2 mg/kg every 3 weeks for 12 weeks) followed by ipilimumab (3 mg/kg every 3 weeks for 9 weeks), but with severe liver dysfunction.
Collapse
Affiliation(s)
- Sadanori Furudate
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Taku Fujimura
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yumi Kambayashi
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takanori Hidaka
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Akira Hashimoto
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Setsuya Aiba
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|