2651
|
|
2652
|
Abstract
PURPOSE OF REVIEW Checkpoint blockade has changed the treatment landscape in non-small cell lung cancer (NSCLC), but single-agent approaches are effective for only a select subset of patients. Here, we will review the evidence for combination immunotherapies in NSCLC and the clinical data evaluating the efficacy of this approach. RECENT FINDINGS Clinical trials evaluating combination PD-1 and CTLA-4 blockade as well as PD-1 in combination with agents targeting IDO1, B7-H3, VEGF, and EGFR show promising results. Additional studies targeting other immune pathways like TIGIT, LAG-3, and cellular therapies are ongoing. Combination immunotherapy has the potential to improve outcomes in NSCLC. Data from early clinical trials is promising and reveals that these agents can be administered together safely without a significant increase in toxicity. Further studies are needed to evaluate their long-term safety and efficacy and to determine appropriate patient selection.
Collapse
Affiliation(s)
| | - Charu Aggarwal
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, USA. .,Hematology-Oncology Division, Department of Medicine, Hospital of the University of Pennsylvania, South Pavilion, Floor 10, 3400 Civic Center Blvd., Philadelphia, PA, 19104, USA.
| |
Collapse
|
2653
|
Scherwitzl I, Hurtado A, Pierce CM, Vogt S, Pampeno C, Meruelo D. Systemically Administered Sindbis Virus in Combination with Immune Checkpoint Blockade Induces Curative Anti-tumor Immunity. MOLECULAR THERAPY-ONCOLYTICS 2018; 9:51-63. [PMID: 29988525 PMCID: PMC6026467 DOI: 10.1016/j.omto.2018.04.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Accepted: 04/28/2018] [Indexed: 12/19/2022]
Abstract
Oncolytic viruses represent a promising form of cancer immunotherapy. We investigated the potential of Sindbis virus (SV) for the treatment of solid tumors expressing the human cancer testis antigen NYESO-1. NYESO-1 is an immunogenic antigen frequently expressed in numerous cancers, such as ovarian cancer. We show that SV expressing the tumor-associated antigen NYESO-1 (SV-NYESO1) acts as an immunostimulatory agent, inducing systemic and rapid lymphocyte activation, leading to a pro-inflammatory environment. SV-NYESO1 treatment combined with anti-programmed death 1 (anti-PD-1) markedly augmented the anti-tumor immunity in mice over the course of treatment, resulting in an avid systemic and intratumoral immune response. This response involved reduced presence of granulocytic myeloid-derived suppressor cells in tumors and an increase in the activation of splenic and tumor-infiltrating T cells. Combined therapy also induced enhanced cytotoxic activity of T cells against NYESO-1-expressing tumors. These results were in line with an observed inverse correlation between T cell activation and tumor growth. Finally, we show that combined therapy resulted in complete clearance of NYESO-1-expressing tumors in vivo and led to long-term protection against recurrences. These findings provide a rationale for clinical studies of SV-NYESO1 combined with immune checkpoint blockade anti-PD-1 to be used in the treatment of NYESO-1-expressing tumors.
Collapse
Affiliation(s)
- Iris Scherwitzl
- Department of Pathology, NYU School of Medicine, New York, NY 10016, USA
| | - Alicia Hurtado
- Department of Pathology, NYU School of Medicine, New York, NY 10016, USA
| | - Carolyn M Pierce
- Department of Pathology, NYU School of Medicine, New York, NY 10016, USA
| | - Sandra Vogt
- Department of Pathology, NYU School of Medicine, New York, NY 10016, USA
| | - Christine Pampeno
- Department of Pathology, NYU School of Medicine, New York, NY 10016, USA
| | - Daniel Meruelo
- Department of Pathology, NYU School of Medicine, New York, NY 10016, USA
| |
Collapse
|
2654
|
Review of diagnostic, prognostic, and predictive biomarkers in melanoma. Clin Exp Metastasis 2018; 35:487-493. [PMID: 29722000 DOI: 10.1007/s10585-018-9892-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 04/16/2018] [Indexed: 12/28/2022]
Abstract
Melanoma is an aggressive cutaneous malignancy with rapidly rising incidence. Diagnosis of controversial melanocytic lesions, correct prognostication of patients, selection of appropriate adjuvant and systemic therapies, and prediction of response to a given therapy remain very real challenges. Despite these challenges, multiple high throughput, nucleic-acid based biomarkers have been developed that can be assayed from histologic tissue specimens. FISH, CGH, Decision-Dx, and other multi-marker assays have been combined to improve overall predictability. This review discusses some of the most promising nucleic acid based assays that can be obtained from tissue specimens to assist with diagnosis, prognostication, and prediction of treatment response.
Collapse
|
2655
|
Gobbini E, Giaj Levra M. Is there a room for immune checkpoint inhibitors in early stage non-small cell lung cancer? J Thorac Dis 2018; 10:S1427-S1437. [PMID: 29951294 DOI: 10.21037/jtd.2018.01.81] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Early non-small cell lung cancer (NSCLC) represents 16% of all new NSCLC at diagnosis with a 5-year survival rate of about 60%. Surgical intervention and adjuvant platinum-based chemotherapy represent the cornerstone treatments, but no significant advances have been achieved since several decades in term of relapse rate reduction or survival improvement. Immunotherapy represents an appealing strategy considering the acceptable toxicity profile but, despite the awesome changing recently introduced in the locally advanced and metastatic setting, its role in early NSCLC is not clear yet. In the past few years, two strategies have been investigated to improve the early NSCLC outcomes eliciting the anti-tumour immune response: tumour vaccines and adoptive cellular therapies. However, none of them provided convincing results. Preclinical and clinical data supported the prognostic role of immune checkpoints in resected NSCLC even if they did not show a clear predictive value for adjuvant treatment. However, some preliminary data about safety and efficacy of neo-adjuvant immune checkpoint inhibitors encourage further investigation of their potential role as monotherapy or as part of a multimodal strategy. Then, even if no significant progress has been done in early NSCLC treatment until today, checkpoint inhibitors can open the door to a new strategy in this setting.
Collapse
Affiliation(s)
- Elisa Gobbini
- Department of Oncology, University of Turin, AOU San Luigi Gonzaga, Orbassano, Italy
| | | |
Collapse
|
2656
|
Knochelmann HM, Dwyer CJ, Bailey SR, Amaya SM, Elston DM, Mazza-McCrann JM, Paulos CM. When worlds collide: Th17 and Treg cells in cancer and autoimmunity. Cell Mol Immunol 2018; 15:458-469. [PMID: 29563615 PMCID: PMC6068176 DOI: 10.1038/s41423-018-0004-4] [Citation(s) in RCA: 372] [Impact Index Per Article: 53.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2017] [Revised: 01/08/2018] [Accepted: 01/09/2018] [Indexed: 12/24/2022] Open
Abstract
The balance between Th17 cells and regulatory T cells (Tregs) has emerged as a prominent factor in regulating autoimmunity and cancer. Th17 cells are vital for host defense against pathogens but have also been implicated in causing autoimmune disorders and cancer, though their role in carcinogenesis is less well understood. Tregs are required for self-tolerance and defense against autoimmunity and often correlate with cancer progression. This review addresses the importance of a functional homeostasis between these two subsets in health and the consequences of its disruption when these forces collide in disease. Importantly, we discuss the ability of Th17 cells to mediate cancer regression in immunotherapy, including adoptive transfer and checkpoint blockade therapy, and the therapeutic possibilities of purposefully offsetting the Th17/Treg balance to treat patients with cancer as well as those with autoimmune diseases.
Collapse
Affiliation(s)
- Hannah M Knochelmann
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA.
- Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston, SC, USA.
| | - Connor J Dwyer
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
- Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston, SC, USA
| | - Stefanie R Bailey
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
- Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston, SC, USA
| | - Sierra M Amaya
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
- Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston, SC, USA
| | - Dirk M Elston
- Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston, SC, USA
| | - Joni M Mazza-McCrann
- Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston, SC, USA
| | - Chrystal M Paulos
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA.
- Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston, SC, USA.
| |
Collapse
|
2657
|
Perioperative Immunotherapy in Muscle-Invasive Bladder Cancer and Upper Tract Urothelial Carcinoma. Urol Clin North Am 2018; 45:287-295. [DOI: 10.1016/j.ucl.2017.12.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
2658
|
Donini C, D'Ambrosio L, Grignani G, Aglietta M, Sangiolo D. Next generation immune-checkpoints for cancer therapy. J Thorac Dis 2018; 10:S1581-S1601. [PMID: 29951308 DOI: 10.21037/jtd.2018.02.79] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The discovery and clinical application of immune-checkpoint inhibitors has dramatically improved the treatments, outcomes and therapeutic concepts in multiple tumor settings. This breakthrough was mainly based on monoclonal antibodies blocking the inhibitory molecule CTLA-4 and or the PD-1/PD-L1 axis, with the aim of counteracting major tumor immune evasion mechanisms. Even acknowledging these important successes, not all the patients benefit from these treatments. Translational and clinical research efforts are ongoing to explore the potentialities of a new generation of immune-modulatory molecules to extend current clinical applications and contrast the unsolved issues of resistance and disease relapse that still affects a considerable rate of patients. New immune-checkpoints, with either stimulatory or inhibitory functions are emerging with key roles in regulating T cell response but also affecting other crucial effectors belonging to the innate immune response (e.g., natural killer). Their therapeutic exploitation, either alone or in strategical combinations, is providing important preclinical results, holding promises currently explored in initial clinical trials. The first results point toward favorable safety profiles with selective hints of activity in challenging settings. Important issues regarding the dose, schedule and rational combinations remain open and data from the clinical studies are needed. Here we provide an overview of the main emerging stimulatory or inhibitory immune-checkpoints exploitable in cancer treatment, briefly reporting their biological function, preclinical activity and preliminary clinical data.
Collapse
Affiliation(s)
- Chiara Donini
- Department of Oncology, University of Torino, Torino, Italy
| | - Lorenzo D'Ambrosio
- Department of Oncology, University of Torino, Torino, Italy.,Division of Medical Oncology, Sarcoma Unit, Candiolo Cancer Institute FPO-IRCCS, Candiolo, Torino, Italy
| | - Giovanni Grignani
- Department of Oncology, University of Torino, Torino, Italy.,Division of Medical Oncology, Sarcoma Unit, Candiolo Cancer Institute FPO-IRCCS, Candiolo, Torino, Italy
| | - Massimo Aglietta
- Department of Oncology, University of Torino, Torino, Italy.,Division of Medical Oncology, Sarcoma Unit, Candiolo Cancer Institute FPO-IRCCS, Candiolo, Torino, Italy.,Division of Medical Oncology, Experimental Cell Therapy, Candiolo Cancer Institute FPO-IRCCS, Candiolo, Torino, Italy
| | - Dario Sangiolo
- Department of Oncology, University of Torino, Torino, Italy.,Division of Medical Oncology, Experimental Cell Therapy, Candiolo Cancer Institute FPO-IRCCS, Candiolo, Torino, Italy
| |
Collapse
|
2659
|
Achard C, Surendran A, Wedge ME, Ungerechts G, Bell J, Ilkow CS. Lighting a Fire in the Tumor Microenvironment Using Oncolytic Immunotherapy. EBioMedicine 2018; 31:17-24. [PMID: 29724655 PMCID: PMC6013846 DOI: 10.1016/j.ebiom.2018.04.020] [Citation(s) in RCA: 116] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 04/20/2018] [Accepted: 04/20/2018] [Indexed: 02/09/2023] Open
Abstract
Oncolytic virus (OV) therapy is potentially a game-changing cancer treatment that has garnered significant interest due to its versatility and multi-modal approaches towards tumor eradication. In the field of cancer immunotherapy, the immunological phenotype of the tumor microenvironment (TME) is an important determinant of disease prognosis and therapeutic success. There is accumulating data that OVs are capable of dramatically altering the TME immune landscape, leading to improved antitumor activity alone or in combination with assorted immune modulators. Herein, we review how OVs disrupt the immunosuppressive TME and can be used strategically to create a "pro-immune" microenvironment that enables and promotes potent, long-lasting host antitumor immune responses.
Collapse
Affiliation(s)
- Carole Achard
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa K1H 8L6, Canada
| | - Abera Surendran
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa K1H 8M5, Canada
| | - Marie-Eve Wedge
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa K1H 8L6, Canada; Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
| | - Guy Ungerechts
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa K1H 8L6, Canada; Department of Medical Oncology and Translational Oncology, National Center for Tumor Diseases (NCT), and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - John Bell
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa K1H 8M5, Canada
| | - Carolina S Ilkow
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa K1H 8M5, Canada.
| |
Collapse
|
2660
|
Tay R, Prelaj A, Califano R. Immune checkpoint blockade for advanced non-small cell lung cancer: challenging clinical scenarios. J Thorac Dis 2018; 10:S1494-S1502. [PMID: 29951301 PMCID: PMC5994491 DOI: 10.21037/jtd.2018.01.80] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 01/08/2018] [Indexed: 12/18/2022]
Abstract
Immune checkpoint blockade has shown anti-tumour activity and improved survival in advanced non-small cell lung cancer (NSCLC). A number of anti-PD-1/PD-L1 and CTLA-4 monoclonal antibody agents have been evaluated in metastatic non-small cell lung cancer. Nivolumab, pembrolizumab and atezolizumab are currently approved for use in clinical practice due to demonstrated improvement in response rate, overall survival (OS) and quality of life (QoL) over standard chemotherapy. We present a series of cases that highlight the clinical challenges that these novel agents present. A review of rare immune-related adverse events (AEs), optimal treatment duration and patient selection will be presented. This series will also address real-life clinical scenarios such as treatment re-challenge and management of immune-related AEs.
Collapse
Affiliation(s)
- Rebecca Tay
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK
| | - Arsela Prelaj
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK
- Department of Radiological, Pathological and Oncological Science, Sapienza University of Rome, Roma, Italy
| | - Raffaele Califano
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK
- Department of Medical Oncology, Manchester University NHS Foundation Trust, Manchester, UK
- Division of Cancer Sciences, University of Manchester, Manchester, UK
| |
Collapse
|
2661
|
Nsengimana J, Laye J, Filia A, O’Shea S, Muralidhar S, Poźniak J, Droop A, Chan M, Walker C, Parkinson L, Gascoyne J, Mell T, Polso M, Jewell R, Randerson-Moor J, Cook GP, Bishop DT, Newton-Bishop J. β-Catenin-mediated immune evasion pathway frequently operates in primary cutaneous melanomas. J Clin Invest 2018; 128:2048-2063. [PMID: 29664013 PMCID: PMC5919828 DOI: 10.1172/jci95351] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 02/27/2018] [Indexed: 12/19/2022] Open
Abstract
Immunotherapy prolongs survival in only a subset of melanoma patients, highlighting the need to better understand the driver tumor microenvironment. We conducted bioinformatic analyses of 703 transcriptomes to probe the immune landscape of primary cutaneous melanomas in a population-ascertained cohort. We identified and validated 6 immunologically distinct subgroups, with the largest having the lowest immune scores and the poorest survival. This poor-prognosis subgroup exhibited expression profiles consistent with β-catenin-mediated failure to recruit CD141+ DCs. A second subgroup displayed an equally bad prognosis when histopathological factors were adjusted for, while 4 others maintained comparable survival profiles. The 6 subgroups were replicated in The Cancer Genome Atlas (TCGA) melanomas, where β-catenin signaling was also associated with low immune scores predominantly related to hypomethylation. The survival benefit of high immune scores was strongest in patients with double-WT tumors for BRAF and NRAS, less strong in BRAF-V600 mutants, and absent in NRAS (codons 12, 13, 61) mutants. In summary, we report evidence for a β-catenin-mediated immune evasion in 42% of melanoma primaries overall and in 73% of those with the worst outcome. We further report evidence for an interaction between oncogenic mutations and host response to melanoma, suggesting that patient stratification will improve immunotherapeutic outcomes.
Collapse
Affiliation(s)
- Jérémie Nsengimana
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, United Kingdom
| | - Jon Laye
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, United Kingdom
| | - Anastasia Filia
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, United Kingdom
- National Heart and Lung Institute, Imperial College, London, United Kingdom
| | - Sally O’Shea
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, United Kingdom
| | - Sathya Muralidhar
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, United Kingdom
| | - Joanna Poźniak
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, United Kingdom
| | - Alastair Droop
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, United Kingdom
- Medical Research Council (MRC) Medical Bioinformatics Centre, University of Leeds, Leeds, United Kingdom
| | - May Chan
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, United Kingdom
| | - Christy Walker
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, United Kingdom
| | - Louise Parkinson
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, United Kingdom
| | - Joanne Gascoyne
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, United Kingdom
| | - Tracey Mell
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, United Kingdom
| | - Minttu Polso
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, United Kingdom
| | - Rosalyn Jewell
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, United Kingdom
- Yorkshire Regional Genetics Service, Leeds Teaching Hospitals NHS Trust, Leeds, United Kingdom
| | | | - Graham P. Cook
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, United Kingdom
| | - D. Timothy Bishop
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, United Kingdom
| | - Julia Newton-Bishop
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
2662
|
Machado I, López-Guerrero JA, Scotlandi K, Picci P, Llombart-Bosch A. Immunohistochemical analysis and prognostic significance of PD-L1, PD-1, and CD8+ tumor-infiltrating lymphocytes in Ewing's sarcoma family of tumors (ESFT). Virchows Arch 2018; 472:815-824. [PMID: 29445891 DOI: 10.1007/s00428-018-2316-2] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 01/10/2018] [Accepted: 01/31/2018] [Indexed: 12/12/2022]
Abstract
Ewing's sarcoma family of tumors (ESFT) are aggressive neoplasms with scant tumor-infiltrating lymphocytes. We analyzed the immunohistochemical (IHC) expression of PD-L1 and PD-1 and their prognostic significance in clinically localized neoplasms in a cohort of 370 ESFT. Slides prepared from tissue microarrays were stained for PD-L1, PD-1, and CD8. Membranous/cytoplasmic staining over 5% of tumor cells was regarded as positive for PD-L1 and PD-1. Prognostic analysis was done considering only clinically localized tumors (n = 217). PD-L1 expression was present in 19% of ESFT, while PD-1 was expressed in 26%. Forty-eight percent of tumors were negative and 12% were positive for both PD-L1 and PD-1. Metastatic tumors displayed higher expression of PD-L1 (p < 0.0001). Histological subtypes were not correlated with PD-L1 or PD-1 positivity. ESFT with elevated proliferation index (Ki-67) were associated with higher PD-L1 expression (p = 0.049). Regarding prognosis, no significant association was found between PD-L1 expression and progression-free survival (PFS) or overall survival (OS), whereas lack of PD-1 expression in tumor cells was correlated with both poor PFS (p = 0.02) and poor OS (p = 0.004). Tumor-infiltrating CD8(+) T lymphocytes were observed in 15.4% of ESFT with informative results (347 tumors). No correlation was found between tumor-infiltrating CD8(+) T lymphocytes and ESFT histological subtypes, tumor location, or PD-1 and PD-L1 expression, nor with PFS (p = 0.473) or OS (p = 0.087). PD-L1 expression was not significantly related to prognosis. PD-1 was expressed in 26% of ESFT tumor cells and may have prognostic and therapeutic implications. CD8 expression in tumor-infiltrating lymphocytes was not related to prognosis.
Collapse
Affiliation(s)
- Isidro Machado
- Pathology Department, Instituto Valenciano de Oncología, Valencia, Spain.
| | | | - Katia Scotlandi
- Laboratory of Experimental Oncology, Istituto Rizzoli, Bologna, Italy
| | - Piero Picci
- Laboratory of Experimental Oncology, Istituto Rizzoli, Bologna, Italy
| | | |
Collapse
|
2663
|
Melanoma of the lower genital tract: Prognostic factors and treatment modalities. Gynecol Oncol 2018; 150:180-189. [PMID: 29728261 DOI: 10.1016/j.ygyno.2018.04.562] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2018] [Revised: 04/16/2018] [Accepted: 04/17/2018] [Indexed: 12/13/2022]
Abstract
Primary melanomas originating from the gynecological tract are rare and aggressive cancers. The vulva is the most frequent site (70%), followed by vagina and more rarely by cervix. The clinical outcome of patients with female genital tract melanoma is very poor, with a 5-year overall survival (OS) of 37-50% for vulvar, 13-32% for vaginal, and approximately 10% for cervical melanoma. In this systematic review, we analyzed the pathogenesis and the different factors influencing the prognosis of melanomas of the lower genital tract, with particular emphasis on biologic variables that may influence new therapeutic approaches. We evaluated the different treatment modalities described in the literature, in order to offer a possible algorithm that may help the clinicians in diagnosing and treating patients with these uncommon malignancies.
Collapse
|
2664
|
Lenormand C, Battistella M, Velter C. Cancérologie cutanée et psoriasis. Ann Dermatol Venereol 2018; 145 Suppl 2:IIS3-IIS22. [PMID: 29699629 DOI: 10.1016/s0151-9638(18)30367-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Affiliation(s)
- C Lenormand
- CHU de Strasbourg, hôpitaux universitaires de Strasbourg, 1, place de l'Höpital, 67000 Strasbourg, France.
| | - M Battistella
- Service de pathologie, hôpital Saint-Louis, université Paris Diderot, 1, avenue Claude-Vellefaux, 75010 Paris
| | - C Velter
- Faculté de médecine, université de Strasbourg et clinique dermatologique, hôpitaux universitaires de Strasbourg, 1, place de l'Höpital, 67091 Strasbourg, France
| |
Collapse
|
2665
|
Dranitsaris G, Zhu X, Adunlin G, Vincent MD. Cost effectiveness vs. affordability in the age of immuno-oncology cancer drugs. Expert Rev Pharmacoecon Outcomes Res 2018; 18:351-357. [PMID: 29681201 DOI: 10.1080/14737167.2018.1467270] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION After years of setback, cancer immunotherapy has begun to yield clinical dividends, which are changing the treatment landscape and offering cancer patients the potential for long-term survival, reduced treatment-related toxicity and improved quality-of-life. Using the immune system to treat cancer is known as 'Immuno-oncology' (IO) and agents are sub-classified by their ability to enhance anti-tumor response or to direct the immune system to attack cancer cells via tumor-associated antigens. Areas covered: Clinical trials have demonstrated the effectiveness of several IO agents in many disease sites such as early and advanced stage melanoma, advanced non-small cell lung cancer, bladder, head and neck, gastric, kidney as well as Hodgkin's lymphoma. Notwithstanding the therapeutic excitement generated for patients and clinicians alike, an important consideration is treatment cost, which can reach more than $US100,000 per patient annually. The cost of the drugs, coupled with high disease prevalence and the ever-expanding number of indications, means the current cost trajectory is untenable for most healthcare systems to sustain. Expert commentary: In this paper, the approved IO drugs and those in clinical development are reviewed. The issue of cost effectiveness vs. affordability is then addressed and suggestions that facilitate patient access and long-term sustainability are presented.
Collapse
Affiliation(s)
| | - Xiaofu Zhu
- b Cross Cancer Institute , Edmonton , Canada
| | - Georges Adunlin
- c School of Pharmacy , Samford University , Birmingham , AL , USA
| | | |
Collapse
|
2666
|
Buglioni S, Melucci E, Sperati F, Pallocca M, Terrenato I, De Nicola F, Goeman F, Casini B, Amoreo CA, Gallo E, Diodoro MG, Pescarmona E, Vici P, Sergi D, Pizzuti L, Di Lauro L, Mazzotta M, Barba M, Fanciulli M, Vitale I, De Maria R, Ciliberto G, Maugeri-Saccà M. The clinical significance of PD-L1 in advanced gastric cancer is dependent on ARID1A mutations and ATM expression. Oncoimmunology 2018; 7:e1457602. [PMID: 30221053 DOI: 10.1080/2162402x.2018.1457602] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 03/15/2018] [Accepted: 03/19/2018] [Indexed: 01/05/2023] Open
Abstract
Whether PD-L1 expression is associated with survival outcomes in gastric cancer (GC) is controversial. The inhibition of the PD-1/PD-L1 pathway is effective against genomically unstable tumors. Hypothesizing that also the clinical significance of PD-L1 might be dependent on the activation of molecular circuits ensuring genomic stability, we evaluated PD-L1 expression in tissue samples from 72 advanced GC patients treated with first-line chemotherapy. Samples were already characterized for DNA damage repair (DDR) component expression (pATM, pChk1, pWee1, γ-H2AX and pRPA2) along with mutations in DDR-linked genes (TP53 and ARID1A). Overall, PD-L1 expression was not associated with progression-free survival (PFS) and overall survival (OS), independently on whether we considered its expression in tumor cells (PD-L1-TCs) or in the immune infiltrate (PD-L1-TILs). In subgroup analysis, positive PD-L1-TC immunostaining was associated with better PFS in patients whose tumors did not carry DDR activation (multivariate Cox: HR 0.34, 95%CI: 0.15-0.76, p = 0.008). This subset (DDRoff) was characterized by negative pATM expression or the presence of ARID1A mutations. Conversely, the relationship between PD-L1-TC expression and PFS was lost in a molecular scenario denoting DDR activation (DDRon), as defined by concomitant pATM expression and ARID1A wild-type form. Surprisingly, while PD-L1-TC expression was associated with better OS in the DDRoff subset (multivariate Cox: HR 0.41, 95% CI: 0.17-0.96, p = 0.039), in the DDRon subgroup we observed an opposite impact on OS (multivariate Cox: HR 2.56, 95%CI: 1.06-6.16, p = 0.036). Thus, PD-L1-TC expression may impact survival outcomes in GC on the basis of the activation/inactivation of genome-safeguarding pathways.
Collapse
Affiliation(s)
- Simonetta Buglioni
- Department of Pathology, "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, Rome, Italy
| | - Elisa Melucci
- Department of Pathology, "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, Rome, Italy
| | - Francesca Sperati
- Biostatistics-Scientific Direction, "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, Rome, Italy
| | - Matteo Pallocca
- SAFU Laboratory, Department of Research, Advanced Diagnostic, and Technological Innovation, "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, Rome, Italy
| | - Irene Terrenato
- Biostatistics-Scientific Direction, "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, Rome, Italy
| | - Francesca De Nicola
- SAFU Laboratory, Department of Research, Advanced Diagnostic, and Technological Innovation, "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, Rome, Italy
| | - Frauke Goeman
- Department of Diagnostic Research and Technological Innovation, Oncogenomic and Epigenetic Unit, "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, Rome, Italy
| | - Beatrice Casini
- Department of Pathology, "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, Rome, Italy
| | - Carla Azzurra Amoreo
- Department of Pathology, "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, Rome, Italy
| | - Enzo Gallo
- Department of Pathology, "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, Rome, Italy
| | - Maria Grazia Diodoro
- Department of Pathology, "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, Rome, Italy
| | - Edoardo Pescarmona
- Department of Pathology, "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, Rome, Italy
| | - Patrizia Vici
- Division of Medical Oncology 2, "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, Rome, Italy
| | - Domenico Sergi
- Division of Medical Oncology 2, "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, Rome, Italy
| | - Laura Pizzuti
- Division of Medical Oncology 2, "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, Rome, Italy
| | - Luigi Di Lauro
- Division of Medical Oncology 2, "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, Rome, Italy
| | - Marco Mazzotta
- Department of Medical Oncology, Medical Oncology Unit, Policlinico Sant'Andrea, Via Di Grotta Rossa 1035/1039, Rome, Italy
| | - Maddalena Barba
- Division of Medical Oncology 2, "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, Rome, Italy
| | - Maurizio Fanciulli
- SAFU Laboratory, Department of Research, Advanced Diagnostic, and Technological Innovation, "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, Rome, Italy
| | - Ilio Vitale
- Cellular Networks and Molecular Therapeutic Targets Unit, Department of Research, Advanced Diagnostics and Technological Innovation, "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, Rome, Italy.,Department of Biology, University of Rome "Tor Vergata", Via della Ricerca Scientifica 1, Rome, Italy
| | - Ruggero De Maria
- Institute of General Pathology, Catholic University of the Sacred Heart and Fondazione Policlinico Universitario Agostino Gemelli, Largo Agostino Gemelli, 10, Rome, Italy
| | - Gennaro Ciliberto
- Scientific Direction, "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, Rome, Italy
| | - Marcello Maugeri-Saccà
- Division of Medical Oncology 2, "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, Rome, Italy
| |
Collapse
|
2667
|
The expression profile of PD-L1 and CD8 + lymphocyte in pituitary adenomas indicating for immunotherapy. J Neurooncol 2018; 139:89-95. [PMID: 29680903 DOI: 10.1007/s11060-018-2844-2] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 03/21/2018] [Indexed: 02/04/2023]
Abstract
BACKGROUND Pituitary adenomas (PAs) are the second most common brain tumors, and mostly are benign tumors. However, there exists subtypes of PAs refractory to common treatments, and need novel therapy. Programmed death 1 (PD-1) blockade has shown durable objective response in a variety of malignancies, and the key predictive markers for this immunotherapy were PD-L1 and CD8+ tumor-infiltrating lymphocyte (TILs) expression. To evaluate the potential immunotherapy for PAs, we investigated the expression of these two immune markers in PAs. METHODS Immunohistochemistry (IHC) was performed to detect the expression of PD-L1 and CD8+ TILs in PAs. The ratio of positive expression of PD-L1 and CD8+ TILs was compared with chi-squared tests among different subtypes of PAs. The association between their expression profile and clinical parameters was analyzed using a chi-squared test, or Fisher's exact probability test when appropriate. RESULTS One hundred and ninety one patients with PAs were retrospectively involved in this study, consisting of 106 non-functioning PAs (NF-PAs, 55.5%), 40 PRL-secreting PAs (PRL-PAs, 20.9%), 31 GH-secreting PAs (GH-PAs, 16.2%), 9 ACTH-secreting PAs (ACTH-PAs, 4.7%) and 5 plurihormonal adenomas (2.6%) respectively. 36.6% of them were PD-L1 positive and 86.9% were CD8+ TILs positive. The positive PD-L1 immunostaining presented more frequently in functioning PAs (58.8%), compared with that (34.3%) in nonfunctioning group (p = 0.000). Moreover, the rates of PD-L1 expression were more associated with increased blood levels of PRL, GH, ACTH and cortisol. Contrastly, positive CD8+ TILs immunostaining was only correlated with elevated blood level of GH. For the analysis of immune markers with pathological results, PD-L1 expression was associated with PRL and GH immunostaining and higher Ki-67 index. But CD8+ TILs was only correlated with PRL immunostaining. CONCLUSION Our results showed that PD-L1 was frequently expressed in functioning PAs with association of aggressive behaviors in PAs. The immunotherapy could be a promising treatment option of PAs.
Collapse
|
2668
|
Brockwell NK, Parker BS. Tumor inherent interferons: Impact on immune reactivity and immunotherapy. Cytokine 2018; 118:42-47. [PMID: 29681426 DOI: 10.1016/j.cyto.2018.04.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2018] [Revised: 04/05/2018] [Accepted: 04/06/2018] [Indexed: 12/21/2022]
Abstract
Immunotherapy has revolutionized cancer treatment, with sustained responses to immune checkpoint inhibitors reported in a number of malignancies. Such therapeutics are now being trialed in aggressive or advanced cancers that are heavily reliant on untargeted therapies, such as triple negative breast cancer. However, responses have been underwhelming to date and are very difficult to predict, leading to an inability to accurately weigh up the benefit-to-risk ratio for their implementation. The tumor immune microenvironment has been closely linked to immunotherapeutic response, with superior responses observed in patients with T cell-inflamed or 'hot' tumors. One class of cytokines, the type I interferons, are a major dictator of tumor immune infiltration and activation. Tumor cell inherent interferon signaling dramatically influences the immune microenvironment and the expression of immune checkpoint proteins, hence regulators and targets of this pathway are candidate biomarkers of immunotherapeutic response. In support of a link between IFN signaling and immunotherapeutic response, the combination of type I interferon inducers with checkpoint immunotherapy has recently been demonstrated critical for a sustained anti-tumor response in aggressive breast cancer models. Here we review evidence that links type I interferons with a hot tumor immune microenvironment, response to checkpoint inhibitors and reduced risk of metastasis that supports their use as biomarkers and therapeutics in oncology.
Collapse
Affiliation(s)
- Natasha K Brockwell
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, Australia
| | - Belinda S Parker
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, Australia.
| |
Collapse
|
2669
|
Haas L, Wiesner T, Obenauf AC. A new era of proactive melanoma therapy: hit hard, hit early. Br J Dermatol 2018; 178:817-820. [DOI: 10.1111/bjd.16347] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- L. Haas
- Research Institute of Molecular Pathology (IMP) Vienna Biocenter (VBC) Campus‐Vienna‐Biocenter 1 1030 Vienna Austria
| | - T. Wiesner
- Department of Dermatology Medical University of Vienna Waehringer Guertel 18‐20 1090 Vienna, Austria
| | - A. C. Obenauf
- Research Institute of Molecular Pathology (IMP) Vienna Biocenter (VBC) Campus‐Vienna‐Biocenter 1 1030 Vienna Austria
| |
Collapse
|
2670
|
|
2671
|
Martin AM, Nirschl CJ, Polanczyk MJ, Bell WR, Nirschl TR, Harris-Bookman S, Phallen J, Hicks J, Martinez D, Ogurtsova A, Xu H, Sullivan LM, Meeker AK, Raabe EH, Cohen KJ, Eberhart CG, Burger PC, Santi M, Taube JM, Pardoll DM, Drake CG, Lim M. PD-L1 expression in medulloblastoma: an evaluation by subgroup. Oncotarget 2018; 9:19177-19191. [PMID: 29721192 PMCID: PMC5922386 DOI: 10.18632/oncotarget.24951] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 03/13/2018] [Indexed: 01/01/2023] Open
Abstract
Background This study evaluated the expression of PD-L1 and markers of immune mediated resistance in human medulloblastoma (MB), the most common malignant pediatric brain tumor. Results Overall levels of PD-L1 in human MB were low; however, some cases demonstrated robust focal expression associated with increased immune infiltrates. The case with highest PD-L1 expression was a sonic hedgehog (SHH) MB. In cell lines, SHH MB, which are low-MYC expressing, demonstrated both constitutive and inducible expression of PD-L1 while those in Group 3/4 that expressed high levels of MYC had only inducible expression. In vitro, IFN-γ robustly stimulated the expression of PD-L1 in all cell lines while radiation induced variable expression. Forced high MYC expression did not significantly alter PD-L1. Methods Human MB tumor samples were evaluated for expression of PD-L1 and immune cell markers in relation to molecular subgroup assignment. PD-L1 expression was functionally analyzed under conditions of interferon gamma (IFN-γ), radiation, and MYC overexpression. Conclusions MB expresses low levels of PD-L1 facilitating immune escape. Importantly, TH1 cytokine stimulation appears to be the most potent inducer of PD-L1 expression in vitro suggesting that an inflamed tumor microenvironment is necessary for PD-1 pathway activation in this tumor.
Collapse
Affiliation(s)
- Allison M Martin
- Johns Hopkins School of Medicine, Sidney Kimmel Cancer Center, Division of Pediatric Oncology, Baltimore, MD, USA
| | - Christopher J Nirschl
- Johns Hopkins School of Medicine, Sidney Kimmel Cancer Center, Division of Cancer Immunology, Baltimore, MD, USA
| | - Magda J Polanczyk
- Johns Hopkins School of Medicine, Sidney Kimmel Cancer Center, Division of Pediatric Oncology, Baltimore, MD, USA
| | - W Robert Bell
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Thomas R Nirschl
- Johns Hopkins School of Medicine, Department of Pathobiology, Baltimore, MD, USA
| | - Sarah Harris-Bookman
- Johns Hopkins School of Medicine, Department of Ophthalmology, Baltimore, MD, USA
| | - Jillian Phallen
- Johns Hopkins School of Medicine, Sidney Kimmel Cancer Center, Division of Cancer Biology, Baltimore, MD, USA
| | - Jessica Hicks
- Johns Hopkins School of Medicine, Department of Pathology, Division of Kidney and Urologic Pathology, Baltimore, MD, USA
| | - Daniel Martinez
- Children's Hospital of Philadelphia, Department of Pathology and Laboratory Medicine, Philadelphia, PA, USA
| | - Aleksandra Ogurtsova
- Johns Hopkins School of Medicine, Department of Dermatology, Division of Dermatologic Pathology and Oral Pathology, Baltimore, MD, USA
| | - Haiying Xu
- Johns Hopkins School of Medicine, Department of Dermatology, Division of Dermatologic Pathology and Oral Pathology, Baltimore, MD, USA
| | | | - Alan K Meeker
- Johns Hopkins School of Medicine, Department of Pathology, Division of Kidney and Urologic Pathology, Baltimore, MD, USA
| | - Eric H Raabe
- Johns Hopkins School of Medicine, Sidney Kimmel Cancer Center, Division of Pediatric Oncology, Baltimore, MD, USA.,Johns Hopkins School of Medicine, Department of Pathology, Division of Neuropathology, Baltimore, MD, USA
| | - Kenneth J Cohen
- Johns Hopkins School of Medicine, Sidney Kimmel Cancer Center, Division of Pediatric Oncology, Baltimore, MD, USA
| | - Charles G Eberhart
- Johns Hopkins School of Medicine, Department of Pathology, Division of Neuropathology, Baltimore, MD, USA
| | - Peter C Burger
- Johns Hopkins School of Medicine, Department of Pathology, Division of Neuropathology, Baltimore, MD, USA
| | - Mariarita Santi
- Children's Hospital of Philadelphia, Department of Pathology and Laboratory Medicine, Philadelphia, PA, USA
| | - Janis M Taube
- Johns Hopkins School of Medicine, Department of Dermatology, Division of Dermatologic Pathology and Oral Pathology, Baltimore, MD, USA
| | - Drew M Pardoll
- Johns Hopkins School of Medicine, Sidney Kimmel Cancer Center, Division of Cancer Immunology, Baltimore, MD, USA
| | - Charles G Drake
- Columbia University Medical Center, Division of Hematology/Oncology, New York, NY, USA
| | - Michael Lim
- Johns Hopkins School of Medicine, Department of Neurosurgery, Division of Neurosurgical Oncology, Baltimore, MD, USA
| |
Collapse
|
2672
|
Zappasodi R, Merghoub T, Wolchok JD. Emerging Concepts for Immune Checkpoint Blockade-Based Combination Therapies. Cancer Cell 2018; 33:581-598. [PMID: 29634946 PMCID: PMC5896787 DOI: 10.1016/j.ccell.2018.03.005] [Citation(s) in RCA: 363] [Impact Index Per Article: 51.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2018] [Revised: 02/13/2018] [Accepted: 03/05/2018] [Indexed: 12/20/2022]
Abstract
Checkpoint blockade has formally demonstrated that reactivating anti-tumor immune responses can regress tumors. However, this only occurs in a fraction of patients. Incorporating these therapies in more powerful combinations is thus a logical next step. Here, we review functional roles of immune checkpoints and molecular determinants of checkpoint-blockade clinical activity. Limited-size T cell-infiltrated tumors, differing substantially from "self," generally respond to checkpoint blockade. Therefore, we propose that reducing tumor burden and increasing tumor immunogenicity are key factors to improve immunotherapy. Lastly, we outline criteria to select proper immunotherapy combination partners and highlight the importance of activity biomarkers for timely treatment optimization.
Collapse
Affiliation(s)
- Roberta Zappasodi
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Taha Merghoub
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| | - Jedd D Wolchok
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Weill Cornell Medicine, New York, NY 10065, USA.
| |
Collapse
|
2673
|
Wrangle JM, Velcheti V, Patel MR, Garrett-Mayer E, Hill EG, Ravenel JG, Miller JS, Farhad M, Anderton K, Lindsey K, Taffaro-Neskey M, Sherman C, Suriano S, Swiderska-Syn M, Sion A, Harris J, Edwards AR, Rytlewski JA, Sanders CM, Yusko EC, Robinson MD, Krieg C, Redmond WL, Egan JO, Rhode PR, Jeng EK, Rock AD, Wong HC, Rubinstein MP. ALT-803, an IL-15 superagonist, in combination with nivolumab in patients with metastatic non-small cell lung cancer: a non-randomised, open-label, phase 1b trial. Lancet Oncol 2018; 19:694-704. [PMID: 29628312 DOI: 10.1016/s1470-2045(18)30148-7] [Citation(s) in RCA: 327] [Impact Index Per Article: 46.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2017] [Revised: 01/31/2018] [Accepted: 02/01/2018] [Indexed: 12/18/2022]
Abstract
BACKGROUND Immunotherapy with PD-1 or PD-L1 blockade fails to induce a response in about 80% of patients with unselected non-small cell lung cancer (NSCLC), and many of those who do initially respond then develop resistance to treatment. Agonists that target the shared interleukin-2 (IL-2) and IL-15Rβγ pathway have induced complete and durable responses in some cancers, but no studies have been done to assess the safety or efficacy of these agonists in combination with anti-PD-1 immunotherapy. We aimed to define the safety, tolerability, and activity of this drug combination in patients with NSCLC. METHODS In this non-randomised, open-label, phase 1b trial, we enrolled patients (aged ≥18 years) with previously treated histologically or cytologically confirmed stage IIIB or IV NSCLC from three academic hospitals in the USA. Key eligibility criteria included measurable disease, eligibility to receive anti-PD-1 immunotherapy, and an Eastern Cooperative Oncology Group performance status of 0 or 1. Patients received the anti-PD-1 monoclonal antibody nivolumab intravenously at 3 mg/kg (then 240 mg when US Food and Drug Administration [FDA]-approved dosing changed) every 14 days (either as new treatment or continued treatment at the time of disease progression) and the IL-15 superagonist ALT-803 subcutaneously once per week on weeks 1-5 of four 6-week cycles for 6 months. ALT-803 was administered at one of four escalating dose concentrations: 6, 10, 15, or 20 μg/kg. The primary endpoint was to define safety and tolerability and to establish a recommended phase 2 dose of ALT-803 in combination with nivolumab. Analyses were per-protocol and included any patients who received at least one dose of study treatment. This trial is registered with ClinicalTrials.gov, number NCT02523469; phase 2 enrolment of patients is ongoing. FINDINGS Between Jan 18, 2016, and June 28, 2017, 23 patients were enrolled and 21 were treated at four dose levels of ALT-803 in combination with nivolumab. Two patients did not receive treatment because of the development of inter-current illness during enrolment, one patient due to leucopenia and one patient due to pulmonary dysfunction. No dose-limiting toxicities were recorded and the maximum tolerated dose was not reached. The most common adverse events were injection-site reactions (in 19 [90%] of 21 patients) and flu-like symptoms (15 [71%]). The most common grade 3 adverse events, occurring in two patients each, were lymphocytopenia and fatigue. A grade 3 myocardial infarction occurred in one patient. No grade 4 or 5 adverse events were recorded. The recommended phase 2 dose of ALT-803 is 20 μg/kg given once per week subcutaneously in combination with 240 mg intravenous nivolumab every 2 weeks. INTERPRETATION ALT-803 in combination with nivolumab can be safely administered in an outpatient setting. The promising clinical activity observed with the addition of ALT-803 to the regimen of patients with PD-1 monoclonal antibody relapsed and refractory disease shows evidence of anti-tumour activity for a new class of agents in NSCLC. FUNDING Altor BioScience (a NantWorks company), National Institutes of Health, and Medical University of South Carolina Hollings Cancer Center.
Collapse
Affiliation(s)
- John M Wrangle
- Department of Medicine, Division of Hematology and Oncology, and Department of Surgery Medical University of South Carolina, Charleston, SC, USA
| | | | - Manish R Patel
- Department of Medicine, Division of Hematology, Oncology, and Transplantation, University of Minnesota, Minneapolis, MN, USA
| | - Elizabeth Garrett-Mayer
- Department of Medicine, Division of Hematology and Oncology, and Department of Surgery Medical University of South Carolina, Charleston, SC, USA
| | - Elizabeth G Hill
- Department of Medicine, Division of Hematology and Oncology, and Department of Surgery Medical University of South Carolina, Charleston, SC, USA
| | - James G Ravenel
- Department of Medicine, Division of Hematology and Oncology, and Department of Surgery Medical University of South Carolina, Charleston, SC, USA
| | - Jeffrey S Miller
- Department of Medicine, Division of Hematology, Oncology, and Transplantation, University of Minnesota, Minneapolis, MN, USA
| | | | - Kate Anderton
- Department of Medicine, Division of Hematology and Oncology, and Department of Surgery Medical University of South Carolina, Charleston, SC, USA
| | - Kathryn Lindsey
- Department of Medicine, Division of Hematology and Oncology, and Department of Surgery Medical University of South Carolina, Charleston, SC, USA
| | - Michele Taffaro-Neskey
- Department of Medicine, Division of Hematology and Oncology, and Department of Surgery Medical University of South Carolina, Charleston, SC, USA
| | - Carol Sherman
- Department of Medicine, Division of Hematology and Oncology, and Department of Surgery Medical University of South Carolina, Charleston, SC, USA
| | - Samantha Suriano
- Department of Medicine, Division of Hematology and Oncology, and Department of Surgery Medical University of South Carolina, Charleston, SC, USA
| | - Marzena Swiderska-Syn
- Department of Medicine, Division of Hematology and Oncology, and Department of Surgery Medical University of South Carolina, Charleston, SC, USA
| | - Amy Sion
- Department of Medicine, Division of Hematology and Oncology, and Department of Surgery Medical University of South Carolina, Charleston, SC, USA
| | - Joni Harris
- Department of Medicine, Division of Hematology and Oncology, and Department of Surgery Medical University of South Carolina, Charleston, SC, USA
| | - Andie R Edwards
- Department of Medicine, Division of Hematology and Oncology, and Department of Surgery Medical University of South Carolina, Charleston, SC, USA
| | | | | | | | - Mark D Robinson
- Institute of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Carsten Krieg
- Department of Medicine, Division of Hematology and Oncology, and Department of Surgery Medical University of South Carolina, Charleston, SC, USA
| | | | | | | | | | | | | | - Mark P Rubinstein
- Department of Medicine, Division of Hematology and Oncology, and Department of Surgery Medical University of South Carolina, Charleston, SC, USA.
| |
Collapse
|
2674
|
Tétu P, Mangana J, Dummer R, Dutriaux C, Beneton N, Dalle S, Meyer N, Oriano B, Michielin O, Lebbe C. Benefit of the nivolumab and ipilimumab combination in pretreated advanced melanoma. Eur J Cancer 2018; 93:147-149. [DOI: 10.1016/j.ejca.2018.01.062] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Accepted: 01/03/2018] [Indexed: 11/16/2022]
|
2675
|
Eisenstein A, Gonzalez EC, Raghunathan R, Xu X, Wu M, McLean EO, McGee J, Ryu B, Alani RM. Emerging Biomarkers in Cutaneous Melanoma. Mol Diagn Ther 2018; 22:203-218. [PMID: 29411301 DOI: 10.1007/s40291-018-0318-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Earlier identification of aggressive melanoma remains a goal in the field of melanoma research. With new targeted and immune therapies that have revolutionized the care of patients with melanoma, the ability to predict progression and monitor or predict response to therapy has become the new focus of research into biomarkers in melanoma. In this review, promising biomarkers are highlighted. These biomarkers have been used to diagnose melanoma as well as predict progression to advanced disease and response to therapy. The biomarkers take various forms, including protein expression at the level of tissue, genetic mutations of cancer cells, and detection of circulating DNA. First, a brief description is provided about the conventional tissue markers used to stage melanoma, including tumor depth. Next, protein biomarkers, which provide both diagnostic and prognostic information, are described. This is followed by a discussion of important genetic mutations, microRNA, and epigenetic modifications that can provide therapeutic and prognostic material. Finally, emerging serologic biomarkers are reviewed, including circulating melanoma cells and exosomes. Overall the goal is to identify biomarkers that aid in the earlier identification and improved treatment of aggressive melanoma.
Collapse
Affiliation(s)
- Anna Eisenstein
- Department of Dermatology, Boston University School of Medicine, 609 Albany Street, Boston, MA, 02118, USA
| | - Estela Chen Gonzalez
- Department of Dermatology, Boston University School of Medicine, 609 Albany Street, Boston, MA, 02118, USA
| | - Rekha Raghunathan
- Department of Dermatology, Boston University School of Medicine, 609 Albany Street, Boston, MA, 02118, USA
| | - Xixi Xu
- Department of Dermatology, Boston University School of Medicine, 609 Albany Street, Boston, MA, 02118, USA
| | - Muzhou Wu
- Department of Dermatology, Boston University School of Medicine, 609 Albany Street, Boston, MA, 02118, USA
| | - Emily O McLean
- Department of Dermatology, Boston University School of Medicine, 609 Albany Street, Boston, MA, 02118, USA
| | - Jean McGee
- Department of Dermatology, Boston University School of Medicine, 609 Albany Street, Boston, MA, 02118, USA
| | - Byungwoo Ryu
- Department of Dermatology, Boston University School of Medicine, 609 Albany Street, Boston, MA, 02118, USA.
| | - Rhoda M Alani
- Department of Dermatology, Boston University School of Medicine, 609 Albany Street, Boston, MA, 02118, USA.
| |
Collapse
|
2676
|
Leonardi GC, Falzone L, Salemi R, Zanghì A, Spandidos DA, McCubrey JA, Candido S, Libra M. Cutaneous melanoma: From pathogenesis to therapy (Review). Int J Oncol 2018; 52:1071-1080. [PMID: 29532857 PMCID: PMC5843392 DOI: 10.3892/ijo.2018.4287] [Citation(s) in RCA: 250] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 02/08/2018] [Indexed: 02/07/2023] Open
Abstract
In less than 10 years, melanoma treatment has been revolutionized with the approval of tyrosine kinase inhibitors and immune checkpoint inhibitors, which have been shown to have a significant impact on the prognosis of patients with melanoma. The early steps of this transformation have taken place in research laboratories. The mitogen‑activated protein kinase (MAPK) pathway, phosphoinositol‑3‑kinase (PI3K) pathway promote the development of melanoma through numerous genomic alterations on different components of these pathways. Moreover, melanoma cells deeply interact with the tumor microenvironment and the immune system. This knowledge has led to the identification of novel therapeutic targets and treatment strategies. In this review, the epidemiological features of cutaneous melanoma along with the biological mechanisms involved in its development and progression are summarized. The current state‑of‑the‑art of advanced stage melanoma treatment strategies and the currently available evidence of the use of predictive and prognostic biomarkers are also discussed.
Collapse
Affiliation(s)
- Giulia C. Leonardi
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania
| | - Luca Falzone
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania
| | - Rossella Salemi
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania
| | - Antonino Zanghì
- Department of Medical and Surgical Sciences and Advanced Technology 'G.F. Ingrassia', University of Catania, 95125 Catania, Italy
| | - Demetrios A. Spandidos
- Laboratory of Clinical Virology, Medical School, University of Crete, 71003 Heraklion, Crete, Greece
| | - James A. McCubrey
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University, Greenville, NC 27858, USA
| | - Saverio Candido
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania
| | - Massimo Libra
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania
- Research Center of Tumor Prevention, Diagnosis and Cure (CRS PreDiCT), University of Catania, 95123 Catania, Italy
| |
Collapse
|
2677
|
Gaiser MR, Bongiorno M, Brownell I. PD-L1 inhibition with avelumab for metastatic Merkel cell carcinoma. Expert Rev Clin Pharmacol 2018; 11:345-359. [PMID: 29478343 PMCID: PMC6360093 DOI: 10.1080/17512433.2018.1445966] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Merkel cell carcinoma (MCC) is a rare and aggressive neuroendocrine skin cancer that lacks durable responses to traditional chemotherapy. Areas covered: After MCC was shown to be an immunogenic tumor, small trials revealed high objective response rates to PD-1/PD-L1 checkpoint inhibitors. The JAVELIN Merkel 200 (NCT02155647) trial tested the use of avelumab, a human IgG1 monoclonal antibody against PD-L1, in metastatic MCC. Avelumab recently became the first approved drug for metastatic MCC. Expert commentary: By conducting broad phase I studies assessing the safety of avelumab and a small phase II study demonstrating efficacy in this rare orphan tumor type, avelumab gained accelerated approval for the treatment of metastatic MCC. Additional studies are needed to determine how the antibody-dependent cellular cytotoxicity (ADCC) competent Fc region of avelumab contributes to disease control. Remaining questions: Longer follow-up will determine the durability of checkpoint blockade in controlling metastatic MCC. Additional studies will assess the utility and safety of adjuvant checkpoint blockade in patients with excised MCC. How to increase response rates by combining PD-1/PD-L1 blockade with other treatment approaches needs to be explored. In addition, treatment options for MCC patients who fail or do not respond to avelumab need to be identified.
Collapse
Affiliation(s)
- Maria Rita Gaiser
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany
| | | | - Isaac Brownell
- Dermatology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, USA
| |
Collapse
|
2678
|
Yip K, Melcher A, Harrington K, Illidge T, Nobes J, Webster A, Smith D, Lorigan P, Nathan P, Larkin J. Pembrolizumab in Combination with Radiotherapy for Metastatic Melanoma - Introducing the PERM Trial. Clin Oncol (R Coll Radiol) 2018; 30:201-203. [PMID: 29402599 DOI: 10.1016/j.clon.2018.01.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 12/12/2017] [Accepted: 12/14/2017] [Indexed: 12/26/2022]
Affiliation(s)
- K Yip
- Ipswich Hospital, Ipswich, UK.
| | - A Melcher
- Institute of Cancer Research, Sutton, London, UK; The Royal Marsden Hospital, Chelsea, London, UK
| | - K Harrington
- Institute of Cancer Research, Sutton, London, UK; The Royal Marsden Hospital, Chelsea, London, UK
| | - T Illidge
- University of Manchester, Manchester, UK; The Christie NHS Foundation Trust, Withington, Manchester, UK
| | - J Nobes
- Norfolk and Norwich University Hospital, Norwich, UK
| | - A Webster
- Radiotherapy Trials Quality Assurance Group, Mount Vernon Cancer Centre, Northwood, UK
| | - D Smith
- The Royal Marsden Hospital, Chelsea, London, UK
| | - P Lorigan
- University of Manchester, Manchester, UK; The Christie NHS Foundation Trust, Withington, Manchester, UK
| | - P Nathan
- Mount Vernon Cancer Centre, Northwood, UK
| | - J Larkin
- The Royal Marsden Hospital, Chelsea, London, UK
| |
Collapse
|
2679
|
Solomon BL, Garrido-Laguna I. Upper gastrointestinal malignancies in 2017: current perspectives and future approaches. Future Oncol 2018; 14:947-962. [PMID: 29542354 PMCID: PMC5925434 DOI: 10.2217/fon-2017-0597] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 01/02/2018] [Indexed: 12/19/2022] Open
Abstract
The advent of immune checkpoint inhibitors (PD-1, PD-L1 and CTLA-4) has resulted in unprecedented long-term remissions of unresectable cancers. The efficacy of checkpoint inhibitors was recently demonstrated in gastrointestinal malignancies with mismatch repair deficiencies (dMMR). Pembrolizumab became the first tissue-agnostic US FDA-approved drug based on the presence of the predictive biomarker dMMR. In addition, the FDA in 2017 approved pembrolizumab for PD-L1-positive advanced gastric cancer in third-line and second-line hepatocellular therapy. Novel treatment strategies such as using anti-carcinoembryonic antigen (CEA) bispecific T cells have led to remarkable responses in microsatellite instability-low colorectal cancer. Other major breakthroughs in treating upper gastrointestinal malignancies in 2017 are discussed.
Collapse
Affiliation(s)
- Benjamin L Solomon
- Department of Internal Medicine (Division of Oncology), University of Utah School of Medicine, Salt Lake City, UT 84103, USA
| | - Ignacio Garrido-Laguna
- Department of Internal Medicine (Division of Oncology), University of Utah School of Medicine, Salt Lake City, UT 84103, USA
- Center for Investigational Therapeutics at Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT 84103, USA
| |
Collapse
|
2680
|
Chatwal MS, Tanvetyanon T. Malignant mesothelioma clinical trial combines immunotherapy drugs. Immunotherapy 2018; 10:341-344. [DOI: 10.2217/imt-2017-0177] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Immunotherapy by checkpoint inhibitor is effective for a number of solid tumors including malignant mesothelioma. Studies utilizing single-agent PD-1 or PD-L1 inhibitor for mesothelioma have reported tumor response rates in approximately 10–20% of patients treated. Given the success of combining these agents with CTLA-4 inhibitor in melanoma, there is a strong rationale to study it in mesothelioma. Recently results from clinical trials investigating this approach have been released. Though limited by small sample size, the studies conclusively demonstrated feasibility and suggested a modestly higher tumor response rate than one would expect from treatment with single-agent PD-1 or PD-L1 inhibitor. Nevertheless, toxicity was also increased. Immunotherapy-related deaths due to encephalitis, renal failure and hepatitis were observed. Further studies are warranted.
Collapse
Affiliation(s)
- Monica S Chatwal
- Division of Medicine, Division of Hematology/Oncology, University of South Florida/H Lee Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Tawee Tanvetyanon
- Thoracic Oncology Department, H. Lee Moffitt Cancer Center, FL 33612, USA
| |
Collapse
|
2681
|
Heymach J, Krilov L, Alberg A, Baxter N, Chang SM, Corcoran RB, Dale W, DeMichele A, Magid Diefenbach CS, Dreicer R, Epstein AS, Gillison ML, Graham DL, Jones J, Ko AH, Lopez AM, Maki RG, Rodriguez-Galindo C, Schilsky RL, Sznol M, Westin SN, Burstein H. Clinical Cancer Advances 2018: Annual Report on Progress Against Cancer From the American Society of Clinical Oncology. J Clin Oncol 2018; 36:1020-1044. [PMID: 29380678 DOI: 10.1200/jco.2017.77.0446] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
A MESSAGE FROM ASCO'S PRESIDENT I remember when ASCO first conceived of publishing an annual report on the most transformative research occurring in cancer care. Thirteen reports later, the progress we have chronicled is remarkable, and this year is no different. The research featured in ASCO's Clinical Cancer Advances 2018 report underscores the impressive gains in our understanding of cancer and in our ability to tailor treatments to tumors' genetic makeup. The ASCO 2018 Advance of the Year, adoptive cell immunotherapy, allows clinicians to genetically reprogram patients' own immune cells to find and attack cancer cells throughout the body. Chimeric antigen receptor (CAR) T-cell therapy-a type of adoptive cell immunotherapy-has led to remarkable results in young patients with acute lymphoblastic leukemia (ALL) and in adults with lymphoma and multiple myeloma. Researchers are also exploring this approach in other types of cancer. This advance would not be possible without robust federal investment in cancer research. The first clinical trial of CAR T-cell therapy in children with ALL was funded, in part, by grants from the National Cancer Institute (NCI), and researchers at the NCI Center for Cancer Research were the first to report on possible CAR T-cell therapy for multiple myeloma. These discoveries follow decades of prior research on immunology and cancer biology, much of which was supported by federal dollars. In fact, many advances that are highlighted in the 2018 Clinical Cancer Advances report were made possible thanks to our nation's support for biomedical research. Funding from the US National Institutes of Health and the NCI helps researchers pursue critical patient care questions and addresses vital, unmet needs that private industry has little incentive to take on. Federally supported cancer research generates the biomedical innovations that fuel the development and availability of new and improved treatments for patients. We need sustained federal research investment to accelerate the discovery of the next generation of cancer treatments. Another major trend in this year's report is progress in precision medicine approaches to treat cancer. Although precision medicine offers promise to people with cancer and their families, that promise is only as good as our ability to make these treatments available to all patients. My presidential theme, "Delivering Discoveries: Expanding the Reach of Precision Medicine," focuses on tackling this formidable challenge so that new targeted therapies are accessible to anyone who faces a cancer diagnosis. By improving access to high-quality care, harnessing big data on patient outcomes from across the globe, and pursuing innovative clinical trials, I am optimistic that we will speed the delivery of these most promising treatments to more patients. Sincerely, Bruce E. Johnson, FASCO ASCO President, 2017 to 2018.
Collapse
Affiliation(s)
- John Heymach
- John Heymach, Maura L. Gillison, and Shannon Neville Westin, The University of Texas MD Anderson Cancer Center, Houston, TX; Lada Krilov and Richard L. Schilsky, American Society of Clinical Oncology, Alexandria; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Anthony Alberg, Medical University of South Carolina, Charleston, SC; Nancy Baxter, St Michael's Hospital, University of Toronto, Toronto, Ontario, Canada; Susan Marina Chang and Andrew H. Ko, University of California, San Francisco; William Dale, City of Hope National Medical Center, Duarte, CA; Ryan Corcoran, Massachusetts General Hospital; Harold Burstein, Dana-Farber Cancer Institute, Boston, MA; Angela DeMichele and Joshua Jones, University of Pennsylvania, Philadelphia, PA; Catherine S. Magid Diefenbach, University of New York; Andrew S. Epstein, Memorial Sloan Kettering Cancer Center, New York; Robert G. Maki, Hofstra-Northwell School of Medicine, Hempstead, NY; David L. Graham, Carolinas Medical Center, Charlotte, NC; Ana Maria Lopez, University of Utah, Salt Lake City, UT; Carlos Rodriguez-Galindo, St Jude Children's Research Hospital, Memphis, TN; and Mario Sznol, Yale University, New Haven, CT
| | - Lada Krilov
- John Heymach, Maura L. Gillison, and Shannon Neville Westin, The University of Texas MD Anderson Cancer Center, Houston, TX; Lada Krilov and Richard L. Schilsky, American Society of Clinical Oncology, Alexandria; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Anthony Alberg, Medical University of South Carolina, Charleston, SC; Nancy Baxter, St Michael's Hospital, University of Toronto, Toronto, Ontario, Canada; Susan Marina Chang and Andrew H. Ko, University of California, San Francisco; William Dale, City of Hope National Medical Center, Duarte, CA; Ryan Corcoran, Massachusetts General Hospital; Harold Burstein, Dana-Farber Cancer Institute, Boston, MA; Angela DeMichele and Joshua Jones, University of Pennsylvania, Philadelphia, PA; Catherine S. Magid Diefenbach, University of New York; Andrew S. Epstein, Memorial Sloan Kettering Cancer Center, New York; Robert G. Maki, Hofstra-Northwell School of Medicine, Hempstead, NY; David L. Graham, Carolinas Medical Center, Charlotte, NC; Ana Maria Lopez, University of Utah, Salt Lake City, UT; Carlos Rodriguez-Galindo, St Jude Children's Research Hospital, Memphis, TN; and Mario Sznol, Yale University, New Haven, CT
| | - Anthony Alberg
- John Heymach, Maura L. Gillison, and Shannon Neville Westin, The University of Texas MD Anderson Cancer Center, Houston, TX; Lada Krilov and Richard L. Schilsky, American Society of Clinical Oncology, Alexandria; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Anthony Alberg, Medical University of South Carolina, Charleston, SC; Nancy Baxter, St Michael's Hospital, University of Toronto, Toronto, Ontario, Canada; Susan Marina Chang and Andrew H. Ko, University of California, San Francisco; William Dale, City of Hope National Medical Center, Duarte, CA; Ryan Corcoran, Massachusetts General Hospital; Harold Burstein, Dana-Farber Cancer Institute, Boston, MA; Angela DeMichele and Joshua Jones, University of Pennsylvania, Philadelphia, PA; Catherine S. Magid Diefenbach, University of New York; Andrew S. Epstein, Memorial Sloan Kettering Cancer Center, New York; Robert G. Maki, Hofstra-Northwell School of Medicine, Hempstead, NY; David L. Graham, Carolinas Medical Center, Charlotte, NC; Ana Maria Lopez, University of Utah, Salt Lake City, UT; Carlos Rodriguez-Galindo, St Jude Children's Research Hospital, Memphis, TN; and Mario Sznol, Yale University, New Haven, CT
| | - Nancy Baxter
- John Heymach, Maura L. Gillison, and Shannon Neville Westin, The University of Texas MD Anderson Cancer Center, Houston, TX; Lada Krilov and Richard L. Schilsky, American Society of Clinical Oncology, Alexandria; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Anthony Alberg, Medical University of South Carolina, Charleston, SC; Nancy Baxter, St Michael's Hospital, University of Toronto, Toronto, Ontario, Canada; Susan Marina Chang and Andrew H. Ko, University of California, San Francisco; William Dale, City of Hope National Medical Center, Duarte, CA; Ryan Corcoran, Massachusetts General Hospital; Harold Burstein, Dana-Farber Cancer Institute, Boston, MA; Angela DeMichele and Joshua Jones, University of Pennsylvania, Philadelphia, PA; Catherine S. Magid Diefenbach, University of New York; Andrew S. Epstein, Memorial Sloan Kettering Cancer Center, New York; Robert G. Maki, Hofstra-Northwell School of Medicine, Hempstead, NY; David L. Graham, Carolinas Medical Center, Charlotte, NC; Ana Maria Lopez, University of Utah, Salt Lake City, UT; Carlos Rodriguez-Galindo, St Jude Children's Research Hospital, Memphis, TN; and Mario Sznol, Yale University, New Haven, CT
| | - Susan Marina Chang
- John Heymach, Maura L. Gillison, and Shannon Neville Westin, The University of Texas MD Anderson Cancer Center, Houston, TX; Lada Krilov and Richard L. Schilsky, American Society of Clinical Oncology, Alexandria; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Anthony Alberg, Medical University of South Carolina, Charleston, SC; Nancy Baxter, St Michael's Hospital, University of Toronto, Toronto, Ontario, Canada; Susan Marina Chang and Andrew H. Ko, University of California, San Francisco; William Dale, City of Hope National Medical Center, Duarte, CA; Ryan Corcoran, Massachusetts General Hospital; Harold Burstein, Dana-Farber Cancer Institute, Boston, MA; Angela DeMichele and Joshua Jones, University of Pennsylvania, Philadelphia, PA; Catherine S. Magid Diefenbach, University of New York; Andrew S. Epstein, Memorial Sloan Kettering Cancer Center, New York; Robert G. Maki, Hofstra-Northwell School of Medicine, Hempstead, NY; David L. Graham, Carolinas Medical Center, Charlotte, NC; Ana Maria Lopez, University of Utah, Salt Lake City, UT; Carlos Rodriguez-Galindo, St Jude Children's Research Hospital, Memphis, TN; and Mario Sznol, Yale University, New Haven, CT
| | - Ryan B Corcoran
- John Heymach, Maura L. Gillison, and Shannon Neville Westin, The University of Texas MD Anderson Cancer Center, Houston, TX; Lada Krilov and Richard L. Schilsky, American Society of Clinical Oncology, Alexandria; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Anthony Alberg, Medical University of South Carolina, Charleston, SC; Nancy Baxter, St Michael's Hospital, University of Toronto, Toronto, Ontario, Canada; Susan Marina Chang and Andrew H. Ko, University of California, San Francisco; William Dale, City of Hope National Medical Center, Duarte, CA; Ryan Corcoran, Massachusetts General Hospital; Harold Burstein, Dana-Farber Cancer Institute, Boston, MA; Angela DeMichele and Joshua Jones, University of Pennsylvania, Philadelphia, PA; Catherine S. Magid Diefenbach, University of New York; Andrew S. Epstein, Memorial Sloan Kettering Cancer Center, New York; Robert G. Maki, Hofstra-Northwell School of Medicine, Hempstead, NY; David L. Graham, Carolinas Medical Center, Charlotte, NC; Ana Maria Lopez, University of Utah, Salt Lake City, UT; Carlos Rodriguez-Galindo, St Jude Children's Research Hospital, Memphis, TN; and Mario Sznol, Yale University, New Haven, CT
| | - William Dale
- John Heymach, Maura L. Gillison, and Shannon Neville Westin, The University of Texas MD Anderson Cancer Center, Houston, TX; Lada Krilov and Richard L. Schilsky, American Society of Clinical Oncology, Alexandria; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Anthony Alberg, Medical University of South Carolina, Charleston, SC; Nancy Baxter, St Michael's Hospital, University of Toronto, Toronto, Ontario, Canada; Susan Marina Chang and Andrew H. Ko, University of California, San Francisco; William Dale, City of Hope National Medical Center, Duarte, CA; Ryan Corcoran, Massachusetts General Hospital; Harold Burstein, Dana-Farber Cancer Institute, Boston, MA; Angela DeMichele and Joshua Jones, University of Pennsylvania, Philadelphia, PA; Catherine S. Magid Diefenbach, University of New York; Andrew S. Epstein, Memorial Sloan Kettering Cancer Center, New York; Robert G. Maki, Hofstra-Northwell School of Medicine, Hempstead, NY; David L. Graham, Carolinas Medical Center, Charlotte, NC; Ana Maria Lopez, University of Utah, Salt Lake City, UT; Carlos Rodriguez-Galindo, St Jude Children's Research Hospital, Memphis, TN; and Mario Sznol, Yale University, New Haven, CT
| | - Angela DeMichele
- John Heymach, Maura L. Gillison, and Shannon Neville Westin, The University of Texas MD Anderson Cancer Center, Houston, TX; Lada Krilov and Richard L. Schilsky, American Society of Clinical Oncology, Alexandria; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Anthony Alberg, Medical University of South Carolina, Charleston, SC; Nancy Baxter, St Michael's Hospital, University of Toronto, Toronto, Ontario, Canada; Susan Marina Chang and Andrew H. Ko, University of California, San Francisco; William Dale, City of Hope National Medical Center, Duarte, CA; Ryan Corcoran, Massachusetts General Hospital; Harold Burstein, Dana-Farber Cancer Institute, Boston, MA; Angela DeMichele and Joshua Jones, University of Pennsylvania, Philadelphia, PA; Catherine S. Magid Diefenbach, University of New York; Andrew S. Epstein, Memorial Sloan Kettering Cancer Center, New York; Robert G. Maki, Hofstra-Northwell School of Medicine, Hempstead, NY; David L. Graham, Carolinas Medical Center, Charlotte, NC; Ana Maria Lopez, University of Utah, Salt Lake City, UT; Carlos Rodriguez-Galindo, St Jude Children's Research Hospital, Memphis, TN; and Mario Sznol, Yale University, New Haven, CT
| | - Catherine S Magid Diefenbach
- John Heymach, Maura L. Gillison, and Shannon Neville Westin, The University of Texas MD Anderson Cancer Center, Houston, TX; Lada Krilov and Richard L. Schilsky, American Society of Clinical Oncology, Alexandria; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Anthony Alberg, Medical University of South Carolina, Charleston, SC; Nancy Baxter, St Michael's Hospital, University of Toronto, Toronto, Ontario, Canada; Susan Marina Chang and Andrew H. Ko, University of California, San Francisco; William Dale, City of Hope National Medical Center, Duarte, CA; Ryan Corcoran, Massachusetts General Hospital; Harold Burstein, Dana-Farber Cancer Institute, Boston, MA; Angela DeMichele and Joshua Jones, University of Pennsylvania, Philadelphia, PA; Catherine S. Magid Diefenbach, University of New York; Andrew S. Epstein, Memorial Sloan Kettering Cancer Center, New York; Robert G. Maki, Hofstra-Northwell School of Medicine, Hempstead, NY; David L. Graham, Carolinas Medical Center, Charlotte, NC; Ana Maria Lopez, University of Utah, Salt Lake City, UT; Carlos Rodriguez-Galindo, St Jude Children's Research Hospital, Memphis, TN; and Mario Sznol, Yale University, New Haven, CT
| | - Robert Dreicer
- John Heymach, Maura L. Gillison, and Shannon Neville Westin, The University of Texas MD Anderson Cancer Center, Houston, TX; Lada Krilov and Richard L. Schilsky, American Society of Clinical Oncology, Alexandria; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Anthony Alberg, Medical University of South Carolina, Charleston, SC; Nancy Baxter, St Michael's Hospital, University of Toronto, Toronto, Ontario, Canada; Susan Marina Chang and Andrew H. Ko, University of California, San Francisco; William Dale, City of Hope National Medical Center, Duarte, CA; Ryan Corcoran, Massachusetts General Hospital; Harold Burstein, Dana-Farber Cancer Institute, Boston, MA; Angela DeMichele and Joshua Jones, University of Pennsylvania, Philadelphia, PA; Catherine S. Magid Diefenbach, University of New York; Andrew S. Epstein, Memorial Sloan Kettering Cancer Center, New York; Robert G. Maki, Hofstra-Northwell School of Medicine, Hempstead, NY; David L. Graham, Carolinas Medical Center, Charlotte, NC; Ana Maria Lopez, University of Utah, Salt Lake City, UT; Carlos Rodriguez-Galindo, St Jude Children's Research Hospital, Memphis, TN; and Mario Sznol, Yale University, New Haven, CT
| | - Andrew S Epstein
- John Heymach, Maura L. Gillison, and Shannon Neville Westin, The University of Texas MD Anderson Cancer Center, Houston, TX; Lada Krilov and Richard L. Schilsky, American Society of Clinical Oncology, Alexandria; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Anthony Alberg, Medical University of South Carolina, Charleston, SC; Nancy Baxter, St Michael's Hospital, University of Toronto, Toronto, Ontario, Canada; Susan Marina Chang and Andrew H. Ko, University of California, San Francisco; William Dale, City of Hope National Medical Center, Duarte, CA; Ryan Corcoran, Massachusetts General Hospital; Harold Burstein, Dana-Farber Cancer Institute, Boston, MA; Angela DeMichele and Joshua Jones, University of Pennsylvania, Philadelphia, PA; Catherine S. Magid Diefenbach, University of New York; Andrew S. Epstein, Memorial Sloan Kettering Cancer Center, New York; Robert G. Maki, Hofstra-Northwell School of Medicine, Hempstead, NY; David L. Graham, Carolinas Medical Center, Charlotte, NC; Ana Maria Lopez, University of Utah, Salt Lake City, UT; Carlos Rodriguez-Galindo, St Jude Children's Research Hospital, Memphis, TN; and Mario Sznol, Yale University, New Haven, CT
| | - Maura L Gillison
- John Heymach, Maura L. Gillison, and Shannon Neville Westin, The University of Texas MD Anderson Cancer Center, Houston, TX; Lada Krilov and Richard L. Schilsky, American Society of Clinical Oncology, Alexandria; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Anthony Alberg, Medical University of South Carolina, Charleston, SC; Nancy Baxter, St Michael's Hospital, University of Toronto, Toronto, Ontario, Canada; Susan Marina Chang and Andrew H. Ko, University of California, San Francisco; William Dale, City of Hope National Medical Center, Duarte, CA; Ryan Corcoran, Massachusetts General Hospital; Harold Burstein, Dana-Farber Cancer Institute, Boston, MA; Angela DeMichele and Joshua Jones, University of Pennsylvania, Philadelphia, PA; Catherine S. Magid Diefenbach, University of New York; Andrew S. Epstein, Memorial Sloan Kettering Cancer Center, New York; Robert G. Maki, Hofstra-Northwell School of Medicine, Hempstead, NY; David L. Graham, Carolinas Medical Center, Charlotte, NC; Ana Maria Lopez, University of Utah, Salt Lake City, UT; Carlos Rodriguez-Galindo, St Jude Children's Research Hospital, Memphis, TN; and Mario Sznol, Yale University, New Haven, CT
| | - David L Graham
- John Heymach, Maura L. Gillison, and Shannon Neville Westin, The University of Texas MD Anderson Cancer Center, Houston, TX; Lada Krilov and Richard L. Schilsky, American Society of Clinical Oncology, Alexandria; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Anthony Alberg, Medical University of South Carolina, Charleston, SC; Nancy Baxter, St Michael's Hospital, University of Toronto, Toronto, Ontario, Canada; Susan Marina Chang and Andrew H. Ko, University of California, San Francisco; William Dale, City of Hope National Medical Center, Duarte, CA; Ryan Corcoran, Massachusetts General Hospital; Harold Burstein, Dana-Farber Cancer Institute, Boston, MA; Angela DeMichele and Joshua Jones, University of Pennsylvania, Philadelphia, PA; Catherine S. Magid Diefenbach, University of New York; Andrew S. Epstein, Memorial Sloan Kettering Cancer Center, New York; Robert G. Maki, Hofstra-Northwell School of Medicine, Hempstead, NY; David L. Graham, Carolinas Medical Center, Charlotte, NC; Ana Maria Lopez, University of Utah, Salt Lake City, UT; Carlos Rodriguez-Galindo, St Jude Children's Research Hospital, Memphis, TN; and Mario Sznol, Yale University, New Haven, CT
| | - Joshua Jones
- John Heymach, Maura L. Gillison, and Shannon Neville Westin, The University of Texas MD Anderson Cancer Center, Houston, TX; Lada Krilov and Richard L. Schilsky, American Society of Clinical Oncology, Alexandria; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Anthony Alberg, Medical University of South Carolina, Charleston, SC; Nancy Baxter, St Michael's Hospital, University of Toronto, Toronto, Ontario, Canada; Susan Marina Chang and Andrew H. Ko, University of California, San Francisco; William Dale, City of Hope National Medical Center, Duarte, CA; Ryan Corcoran, Massachusetts General Hospital; Harold Burstein, Dana-Farber Cancer Institute, Boston, MA; Angela DeMichele and Joshua Jones, University of Pennsylvania, Philadelphia, PA; Catherine S. Magid Diefenbach, University of New York; Andrew S. Epstein, Memorial Sloan Kettering Cancer Center, New York; Robert G. Maki, Hofstra-Northwell School of Medicine, Hempstead, NY; David L. Graham, Carolinas Medical Center, Charlotte, NC; Ana Maria Lopez, University of Utah, Salt Lake City, UT; Carlos Rodriguez-Galindo, St Jude Children's Research Hospital, Memphis, TN; and Mario Sznol, Yale University, New Haven, CT
| | - Andrew H Ko
- John Heymach, Maura L. Gillison, and Shannon Neville Westin, The University of Texas MD Anderson Cancer Center, Houston, TX; Lada Krilov and Richard L. Schilsky, American Society of Clinical Oncology, Alexandria; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Anthony Alberg, Medical University of South Carolina, Charleston, SC; Nancy Baxter, St Michael's Hospital, University of Toronto, Toronto, Ontario, Canada; Susan Marina Chang and Andrew H. Ko, University of California, San Francisco; William Dale, City of Hope National Medical Center, Duarte, CA; Ryan Corcoran, Massachusetts General Hospital; Harold Burstein, Dana-Farber Cancer Institute, Boston, MA; Angela DeMichele and Joshua Jones, University of Pennsylvania, Philadelphia, PA; Catherine S. Magid Diefenbach, University of New York; Andrew S. Epstein, Memorial Sloan Kettering Cancer Center, New York; Robert G. Maki, Hofstra-Northwell School of Medicine, Hempstead, NY; David L. Graham, Carolinas Medical Center, Charlotte, NC; Ana Maria Lopez, University of Utah, Salt Lake City, UT; Carlos Rodriguez-Galindo, St Jude Children's Research Hospital, Memphis, TN; and Mario Sznol, Yale University, New Haven, CT
| | - Ana Maria Lopez
- John Heymach, Maura L. Gillison, and Shannon Neville Westin, The University of Texas MD Anderson Cancer Center, Houston, TX; Lada Krilov and Richard L. Schilsky, American Society of Clinical Oncology, Alexandria; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Anthony Alberg, Medical University of South Carolina, Charleston, SC; Nancy Baxter, St Michael's Hospital, University of Toronto, Toronto, Ontario, Canada; Susan Marina Chang and Andrew H. Ko, University of California, San Francisco; William Dale, City of Hope National Medical Center, Duarte, CA; Ryan Corcoran, Massachusetts General Hospital; Harold Burstein, Dana-Farber Cancer Institute, Boston, MA; Angela DeMichele and Joshua Jones, University of Pennsylvania, Philadelphia, PA; Catherine S. Magid Diefenbach, University of New York; Andrew S. Epstein, Memorial Sloan Kettering Cancer Center, New York; Robert G. Maki, Hofstra-Northwell School of Medicine, Hempstead, NY; David L. Graham, Carolinas Medical Center, Charlotte, NC; Ana Maria Lopez, University of Utah, Salt Lake City, UT; Carlos Rodriguez-Galindo, St Jude Children's Research Hospital, Memphis, TN; and Mario Sznol, Yale University, New Haven, CT
| | - Robert G Maki
- John Heymach, Maura L. Gillison, and Shannon Neville Westin, The University of Texas MD Anderson Cancer Center, Houston, TX; Lada Krilov and Richard L. Schilsky, American Society of Clinical Oncology, Alexandria; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Anthony Alberg, Medical University of South Carolina, Charleston, SC; Nancy Baxter, St Michael's Hospital, University of Toronto, Toronto, Ontario, Canada; Susan Marina Chang and Andrew H. Ko, University of California, San Francisco; William Dale, City of Hope National Medical Center, Duarte, CA; Ryan Corcoran, Massachusetts General Hospital; Harold Burstein, Dana-Farber Cancer Institute, Boston, MA; Angela DeMichele and Joshua Jones, University of Pennsylvania, Philadelphia, PA; Catherine S. Magid Diefenbach, University of New York; Andrew S. Epstein, Memorial Sloan Kettering Cancer Center, New York; Robert G. Maki, Hofstra-Northwell School of Medicine, Hempstead, NY; David L. Graham, Carolinas Medical Center, Charlotte, NC; Ana Maria Lopez, University of Utah, Salt Lake City, UT; Carlos Rodriguez-Galindo, St Jude Children's Research Hospital, Memphis, TN; and Mario Sznol, Yale University, New Haven, CT
| | - Carlos Rodriguez-Galindo
- John Heymach, Maura L. Gillison, and Shannon Neville Westin, The University of Texas MD Anderson Cancer Center, Houston, TX; Lada Krilov and Richard L. Schilsky, American Society of Clinical Oncology, Alexandria; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Anthony Alberg, Medical University of South Carolina, Charleston, SC; Nancy Baxter, St Michael's Hospital, University of Toronto, Toronto, Ontario, Canada; Susan Marina Chang and Andrew H. Ko, University of California, San Francisco; William Dale, City of Hope National Medical Center, Duarte, CA; Ryan Corcoran, Massachusetts General Hospital; Harold Burstein, Dana-Farber Cancer Institute, Boston, MA; Angela DeMichele and Joshua Jones, University of Pennsylvania, Philadelphia, PA; Catherine S. Magid Diefenbach, University of New York; Andrew S. Epstein, Memorial Sloan Kettering Cancer Center, New York; Robert G. Maki, Hofstra-Northwell School of Medicine, Hempstead, NY; David L. Graham, Carolinas Medical Center, Charlotte, NC; Ana Maria Lopez, University of Utah, Salt Lake City, UT; Carlos Rodriguez-Galindo, St Jude Children's Research Hospital, Memphis, TN; and Mario Sznol, Yale University, New Haven, CT
| | - Richard L Schilsky
- John Heymach, Maura L. Gillison, and Shannon Neville Westin, The University of Texas MD Anderson Cancer Center, Houston, TX; Lada Krilov and Richard L. Schilsky, American Society of Clinical Oncology, Alexandria; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Anthony Alberg, Medical University of South Carolina, Charleston, SC; Nancy Baxter, St Michael's Hospital, University of Toronto, Toronto, Ontario, Canada; Susan Marina Chang and Andrew H. Ko, University of California, San Francisco; William Dale, City of Hope National Medical Center, Duarte, CA; Ryan Corcoran, Massachusetts General Hospital; Harold Burstein, Dana-Farber Cancer Institute, Boston, MA; Angela DeMichele and Joshua Jones, University of Pennsylvania, Philadelphia, PA; Catherine S. Magid Diefenbach, University of New York; Andrew S. Epstein, Memorial Sloan Kettering Cancer Center, New York; Robert G. Maki, Hofstra-Northwell School of Medicine, Hempstead, NY; David L. Graham, Carolinas Medical Center, Charlotte, NC; Ana Maria Lopez, University of Utah, Salt Lake City, UT; Carlos Rodriguez-Galindo, St Jude Children's Research Hospital, Memphis, TN; and Mario Sznol, Yale University, New Haven, CT
| | - Mario Sznol
- John Heymach, Maura L. Gillison, and Shannon Neville Westin, The University of Texas MD Anderson Cancer Center, Houston, TX; Lada Krilov and Richard L. Schilsky, American Society of Clinical Oncology, Alexandria; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Anthony Alberg, Medical University of South Carolina, Charleston, SC; Nancy Baxter, St Michael's Hospital, University of Toronto, Toronto, Ontario, Canada; Susan Marina Chang and Andrew H. Ko, University of California, San Francisco; William Dale, City of Hope National Medical Center, Duarte, CA; Ryan Corcoran, Massachusetts General Hospital; Harold Burstein, Dana-Farber Cancer Institute, Boston, MA; Angela DeMichele and Joshua Jones, University of Pennsylvania, Philadelphia, PA; Catherine S. Magid Diefenbach, University of New York; Andrew S. Epstein, Memorial Sloan Kettering Cancer Center, New York; Robert G. Maki, Hofstra-Northwell School of Medicine, Hempstead, NY; David L. Graham, Carolinas Medical Center, Charlotte, NC; Ana Maria Lopez, University of Utah, Salt Lake City, UT; Carlos Rodriguez-Galindo, St Jude Children's Research Hospital, Memphis, TN; and Mario Sznol, Yale University, New Haven, CT
| | - Shannon Neville Westin
- John Heymach, Maura L. Gillison, and Shannon Neville Westin, The University of Texas MD Anderson Cancer Center, Houston, TX; Lada Krilov and Richard L. Schilsky, American Society of Clinical Oncology, Alexandria; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Anthony Alberg, Medical University of South Carolina, Charleston, SC; Nancy Baxter, St Michael's Hospital, University of Toronto, Toronto, Ontario, Canada; Susan Marina Chang and Andrew H. Ko, University of California, San Francisco; William Dale, City of Hope National Medical Center, Duarte, CA; Ryan Corcoran, Massachusetts General Hospital; Harold Burstein, Dana-Farber Cancer Institute, Boston, MA; Angela DeMichele and Joshua Jones, University of Pennsylvania, Philadelphia, PA; Catherine S. Magid Diefenbach, University of New York; Andrew S. Epstein, Memorial Sloan Kettering Cancer Center, New York; Robert G. Maki, Hofstra-Northwell School of Medicine, Hempstead, NY; David L. Graham, Carolinas Medical Center, Charlotte, NC; Ana Maria Lopez, University of Utah, Salt Lake City, UT; Carlos Rodriguez-Galindo, St Jude Children's Research Hospital, Memphis, TN; and Mario Sznol, Yale University, New Haven, CT
| | - Harold Burstein
- John Heymach, Maura L. Gillison, and Shannon Neville Westin, The University of Texas MD Anderson Cancer Center, Houston, TX; Lada Krilov and Richard L. Schilsky, American Society of Clinical Oncology, Alexandria; Robert Dreicer, University of Virginia School of Medicine, Charlottesville, VA; Anthony Alberg, Medical University of South Carolina, Charleston, SC; Nancy Baxter, St Michael's Hospital, University of Toronto, Toronto, Ontario, Canada; Susan Marina Chang and Andrew H. Ko, University of California, San Francisco; William Dale, City of Hope National Medical Center, Duarte, CA; Ryan Corcoran, Massachusetts General Hospital; Harold Burstein, Dana-Farber Cancer Institute, Boston, MA; Angela DeMichele and Joshua Jones, University of Pennsylvania, Philadelphia, PA; Catherine S. Magid Diefenbach, University of New York; Andrew S. Epstein, Memorial Sloan Kettering Cancer Center, New York; Robert G. Maki, Hofstra-Northwell School of Medicine, Hempstead, NY; David L. Graham, Carolinas Medical Center, Charlotte, NC; Ana Maria Lopez, University of Utah, Salt Lake City, UT; Carlos Rodriguez-Galindo, St Jude Children's Research Hospital, Memphis, TN; and Mario Sznol, Yale University, New Haven, CT
| |
Collapse
|
2682
|
Sosa A, Lopez Cadena E, Simon Olive C, Karachaliou N, Rosell R. Clinical assessment of immune-related adverse events. Ther Adv Med Oncol 2018; 10:1758835918764628. [PMID: 29623110 PMCID: PMC5882039 DOI: 10.1177/1758835918764628] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 02/15/2018] [Indexed: 12/18/2022] Open
Abstract
Immunotherapy through checkpoint inhibitors is now standard practice for a growing number of cancer types, supported by overall improvement of clinical outcomes and better tolerance. One anti-CTLA-4 antibody (ipilimumab), two anti-PD-1 antibodies (pembrolizumab and nivolumab) and three anti-PD-L1 antibodies (atezolizumab, avelumab and durvalumab) have been approved for clear benefits across diverse trials. Adverse events of an immune nature associated with these agents frequently affect the skin, colon, endocrine glands, lungs and liver. Most of these effects are mild and can be managed through transient immunosuppression with corticosteroids, but high-grade events often require hospitalization and specialized treatment. However, since immunotherapy is recent, physicians with clinical experience in the diagnosis and management of immune toxicities are frequently those who actively participated in trials, but many practicing oncologists are still not familiarized with the assessment of these events. This review focuses on the incidence, diagnostic assessment and recommended management of the most relevant immune-related adverse events.
Collapse
Affiliation(s)
- Aaron Sosa
- Department of Medical Oncology, Hospital Universitari Sagrat Cor, Viladomat 288, Barcelona 08029, Spain
| | - Esther Lopez Cadena
- Department of Pneumonology, Hospital Universitari Sagrat Cor, Barcelona, Spain
| | | | - Niki Karachaliou
- Department of Medical Oncology, Hospital Universitari Sagrat Cor, Barcelona, Spain
| | - Rafael Rosell
- Department of Medical Oncology, Dr Rosell Oncology Institute, Barcelona, Spain
| |
Collapse
|
2683
|
Dahlén E, Veitonmäki N, Norlén P. Bispecific antibodies in cancer immunotherapy. Ther Adv Vaccines Immunother 2018; 6:3-17. [PMID: 29998217 DOI: 10.1177/2515135518763280] [Citation(s) in RCA: 152] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 02/07/2018] [Indexed: 12/29/2022] Open
Abstract
Following the clinical success of immune checkpoint antibodies targeting CTLA-4, PD-1 or PD-L1 in cancer treatment, bispecific antibodies are now emerging as a growing class of immunotherapies with potential to further improve clinical efficacy and safety. We describe three classes of immunotherapeutic bispecific antibodies: (a) cytotoxic effector cell redirectors; (b) tumor-targeted immunomodulators; and (c) dual immunomodulators. Cytotoxic effector cell redirectors are dominated by T-cell redirecting compounds, bispecific compounds engaging a tumor-associated antigen and the T-cell receptor/CD3 complex, thereby redirecting T-cell cytotoxicity to malignant cells. This is the most established class of bispecific immunotherapies, with two compounds having reached the market and numerous compounds in clinical development. Tumor-targeted immunomodulators are bispecific compounds binding to a tumor-associated antigen and an immunomodulating receptor, such as CD40 or 4-1BB. Such compounds are usually designed to be inactive until binding the tumor antigen, thereby localizing immune stimulation to the tumor environment, while minimizing immune activation elsewhere. This is expected to induce powerful activation of tumor-specific T cells with reduced risk of immune-related adverse events. Finally, dual immunomodulators are bispecific compounds that bind two distinct immunomodulating targets, often combining targeting of PD-1 or PD-L1 with that of LAG-3 or TIM-3. The rationale is to induce superior tumor immunity compared to monospecific antibodies to the same targets. In this review, we describe each of these classes of bispecific antibodies, and present examples of compounds in development.
Collapse
Affiliation(s)
- Eva Dahlén
- Alligator Bioscience, 22381 Lund, Sweden
| | | | - Per Norlén
- Alligator Bioscience, 22381 Lund, Sweden
| |
Collapse
|
2684
|
Wieder T, Eigentler T, Brenner E, Röcken M. Immune checkpoint blockade therapy. J Allergy Clin Immunol 2018; 142:1403-1414. [PMID: 29596939 DOI: 10.1016/j.jaci.2018.02.042] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 02/09/2018] [Accepted: 02/27/2018] [Indexed: 12/30/2022]
Abstract
Immune checkpoints are accessory molecules that either promote or inhibit T-cell activation. Two inhibitory molecules, cytotoxic T-lymphocyte antigen 4 (CTLA-4) and programmed cell death protein 1 (PD-1), got high attention, as inhibition of CTLA-4 or PD-1 signaling provides the first immune therapy that significantly improves the survival of patients with metastatic solid cancers. Inhibition of CTLA-4 or PD-1 was first studied in and approved for patients with metastatic melanoma. Blocking immune checkpoints is also efficient in non-small-cell lung cancer, renal cell cancers, hypermutated gastrointestinal cancers, and others. Immune responses, whether directed against infections or against tumors, are divided into 2 phases: an initiation phase and an activation phase, where the immune system recognizes a danger signal and becomes activated by innate signals to fight the danger. This reaction is fundamental for the control of infections and cancer, but needs to be turned off once the danger is controlled, because persistence of this activation ultimately causes severe tissue damage. Therefore, each activation of the immune system is followed by a termination phase, where endogenous immune suppressor molecules arrest immune responses to prevent harmful damage. In the case of cancer immune therapies, therapeutic approaches classically enhanced the initiation and activation of immune responses to increase the emergence and the efficacy of cytotoxic T lymphocytes (CTL) against cancers. In sharp contrast, immune checkpoint blockade focuses on the termination of immune responses by inhibiting immune suppressor molecules. It thus prevents the termination of immune responses or even awakes those CTLs that became exhausted during an immune response. Therefore, blocking negatively regulating immune checkpoints restores the capacity of exhausted CTL to kill the cancer they infiltrate. In addition, they drive surviving cancer cells into a still poorly defined state of dormancy. As the therapy also awakes self-reactive CTL, one downside of the therapy is the induction of organ-specific autoimmune diseases. The second downside is the exorbitant drug price that withdraws patients in need from a therapy that was developed by academic research, which impairs further academic treatment development and financially charges the public health system.
Collapse
Affiliation(s)
- Thomas Wieder
- Department of Dermatology, Eberhard Karls University, Tübingen, Germany
| | - Thomas Eigentler
- Department of Dermatology, Eberhard Karls University, Tübingen, Germany
| | - Ellen Brenner
- Department of Dermatology, Eberhard Karls University, Tübingen, Germany
| | - Martin Röcken
- Department of Dermatology, Eberhard Karls University, Tübingen, Germany.
| |
Collapse
|
2685
|
Combination nivolumab and ipilimumab or nivolumab alone in melanoma brain metastases: a multicentre randomised phase 2 study. Lancet Oncol 2018; 19:672-681. [PMID: 29602646 DOI: 10.1016/s1470-2045(18)30139-6] [Citation(s) in RCA: 734] [Impact Index Per Article: 104.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 01/29/2018] [Accepted: 01/30/2018] [Indexed: 11/23/2022]
Abstract
BACKGROUND Nivolumab monotherapy and combination nivolumab plus ipilimumab increase proportions of patients achieving a response and survival versus ipilimumab in patients with metastatic melanoma; however, efficacy in active brain metastases is unknown. We aimed to establish the efficacy and safety of nivolumab alone or in combination with ipilimumab in patients with active melanoma brain metastases. METHODS This multicentre open-label randomised phase 2 trial was done at four sites in Australia, in three cohorts of immunotherapy-naive patients aged 18 years or older with melanoma brain metastases. Patients with asymptomatic brain metastases with no previous local brain therapy were randomly assigned using the biased coin minimisation method, stratified by site, in a 30:24 ratio (after a safety run-in of six patients) to cohort A (nivolumab plus ipilimumab) or cohort B (nivolumab). Patients with brain metastases in whom local therapy had failed, or who had neurological symptoms, or leptomeningeal disease were enrolled in non-randomised cohort C (nivolumab). Patients in cohort A received intravenous nivolumab 1 mg/kg combined with ipilimumab 3 mg/kg every 3 weeks for four doses, then nivolumab 3 mg/kg every 2 weeks; patients in cohort B or cohort C received intravenous nivolumab 3 mg/kg every 2 weeks. The primary endpoint was intracranial response from week 12. Primary and safety analyses were done on an intention-to-treat basis in all patients who received at least one dose of the study drug. This trial is registered with ClinicalTrials.gov, number NCT02374242, and is ongoing for the final survival analysis. FINDINGS Between Nov 4, 2014, and April 21, 2017, 79 patients were enrolled; 36 in cohort A, 27 in cohort B, and 16 in cohort C. One patient in cohort A and two in cohort B were found to be ineligible and excluded from the study before receiving the study drug. At the data cutoff (Aug 28, 2017), with a median follow up of 17 months (IQR 8-25), intracranial responses were achieved by 16 (46%; 95% CI 29-63) of 35 patients in cohort A, five (20%; 7-41) of 25 in cohort B, and one (6%; 0-30) of 16 in cohort C. Intracranial complete responses occurred in six (17%) patients in cohort A, three (12%) in cohort B, and none in cohort C. Treatment-related adverse events occurred in 34 (97%) of 35 patients in cohort A, 17 (68%) of 25 in cohort B, and eight (50%) of 16 in cohort C. Grade 3 or 4 treatment-related adverse events occurred in 19 (54%) patients in cohort A, four (16%) in cohort B, and two (13%) in cohort C. No treatment-related deaths occurred. INTERPRETATION Nivolumab combined with ipilimumab and nivolumab monotherapy are active in melanoma brain metastases. A high proportion of patients achieved an intracranial response with the combination. Thus, nivolumab combined with ipilimumab should be considered as a first-line therapy for patients with asymptomatic untreated brain metastases. FUNDING Melanoma Institute Australia and Bristol-Myers Squibb.
Collapse
|
2686
|
Inhibition of Enhancer of zeste homolog 2 (EZH2) induces natural killer cell-mediated eradication of hepatocellular carcinoma cells. Proc Natl Acad Sci U S A 2018; 115:E3509-E3518. [PMID: 29581297 DOI: 10.1073/pnas.1802691115] [Citation(s) in RCA: 115] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Natural killer (NK) cell-mediated tumor cell eradication could inhibit tumor initiation and progression. However, the factors that regulate NK cell-mediated cancer cell eradication remain unclear. We determined that hepatocellular carcinoma (HCC) cells exhibit transcriptional down-regulation of NK group 2D (NKG2D) ligands and are largely resistant to NK cell-mediated eradication. Because the down-regulation of NKG2D ligands occurred at the transcriptional level, we tested 32 chemical inhibitors of epigenetic regulators for their ability to re-express NKG2D ligands and enhance HCC cell eradication by NK cells and found that Enhancer of zeste homolog 2 (EZH2) was a transcriptional repressor of NKG2D ligands. The inhibition of EZH2 by small-molecule inhibitors or genetic means enhanced HCC cell eradication by NK cells in a NKG2D ligand-dependent manner. Collectively, these results demonstrate that EZH2 inhibition enhances HCC eradication by NK cells and that EZH2 functions, in part, as an oncogene by inhibiting immune response.
Collapse
|
2687
|
Expression and Targeting of Tumor Markers in Gelfoam ® Histoculture: Potential Individualized Assays for Immuno-Oncology. Methods Mol Biol 2018. [PMID: 29572791 DOI: 10.1007/978-1-4939-7745-1_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Tumor-specific antigens are important in the study of tumor biology, tumor diagnosis, and prognosis and as targets for tumor therapy. This chapter reviews patient colon, breast, and ovarian tumors in 3-dimensional Gelfoam® histoculture maintaining in vivo-like expression of the important tumor antigens, for example TAG-72 and CEA. We have also reviewed that fluorescent antibodies can target tumors in Gelfoam® histoculture, thereby providing an assay for individual patients for sensitivity to therapeutic antibodies which have become so important in immuno-oncology and other cancer therapies.
Collapse
|
2688
|
Abstract
PURPOSE OF REVIEW Prognosis of patients with advanced melanoma is dismal with a median overall survival of about 8 months and 5-year overall survival from a diagnosis of metastatic disease of roughly 10%. However, immune checkpoint inhibitors have brought indispensable benefits to melanoma patients. Here we will review the recent clinical efficacy and adverse events of immune checkpoint inhibitors for melanoma patients. RECENT FINDINGS The immune checkpoint inhibitors increase confirmed objective response and prolong progression-free and overall survival of the afflicted patients in association with maintaining their quality of life. Although diverse immune-related adverse events occur, most of them are manageable by appropriate immunomodulating agents. Clinical efficacy of immune checkpoint inhibitors continues even after discontinuation of drugs. Compared with conventional therapeutic options, the immune checkpoint inhibitors appear to prolong the survival of patients with advanced melanoma. Further clinical trials are warranted to determine whether their combinatory use with other treatment options may augment benefits or not.
Collapse
|
2689
|
Abstract
The release of negative regulators of immune activation (immune checkpoints) that limit antitumor responses has resulted in unprecedented rates of long-lasting tumor responses in patients with a variety of cancers. This can be achieved by antibodies blocking the cytotoxic T lymphocyte-associated protein 4 (CTLA-4) or the programmed cell death 1 (PD-1) pathway, either alone or in combination. The main premise for inducing an immune response is the preexistence of antitumor T cells that were limited by specific immune checkpoints. Most patients who have tumor responses maintain long-lasting disease control, yet one-third of patients relapse. Mechanisms of acquired resistance are currently poorly understood, but evidence points to alterations that converge on the antigen presentation and interferon-γ signaling pathways. New-generation combinatorial therapies may overcome resistance mechanisms to immune checkpoint therapy.
Collapse
Affiliation(s)
- Antoni Ribas
- Department of Medicine, Division of Hematology-Oncology; Department of Surgery, Division of Surgical Oncology; and Department of Molecular and Medical Pharmacology, Jonsson Comprehensive Cancer Center and Parker Institute for Cancer Immunotherapy, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| | - Jedd D Wolchok
- Department of Medicine, Ludwig Center and Parker Institute for Cancer Immunotherapy at Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA. .,Weill Cornell Medical and Graduate Colleges, New York, NY 10065, USA
| |
Collapse
|
2690
|
Mahmood SS, Fradley MG, Cohen JV, Nohria A, Reynolds KL, Heinzerling LM, Sullivan RJ, Damrongwatanasuk R, Chen CL, Gupta D, Kirchberger MC, Awadalla M, Hassan MZO, Moslehi JJ, Shah SP, Ganatra S, Thavendiranathan P, Lawrence DP, Groarke JD, Neilan TG. Myocarditis in Patients Treated With Immune Checkpoint Inhibitors. J Am Coll Cardiol 2018; 71:1755-1764. [PMID: 29567210 DOI: 10.1016/j.jacc.2018.02.037] [Citation(s) in RCA: 1056] [Impact Index Per Article: 150.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 02/08/2018] [Accepted: 02/08/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND Myocarditis is an uncommon, but potentially fatal, toxicity of immune checkpoint inhibitors (ICI). Myocarditis after ICI has not been well characterized. OBJECTIVES The authors sought to understand the presentation and clinical course of ICI-associated myocarditis. METHODS After observation of sporadic ICI-associated myocarditis cases, the authors created a multicenter registry with 8 sites. From November 2013 to July 2017, there were 35 patients with ICI-associated myocarditis, who were compared to a random sample of 105 ICI-treated patients without myocarditis. Covariates of interest were extracted from medical records including the occurrence of major adverse cardiac events (MACE), defined as the composite of cardiovascular death, cardiogenic shock, cardiac arrest, and hemodynamically significant complete heart block. RESULTS The prevalence of myocarditis was 1.14% with a median time of onset of 34 days after starting ICI (interquartile range: 21 to 75 days). Cases were 65 ± 13 years of age, 29% were female, and 54% had no other immune-related side effects. Relative to controls, combination ICI (34% vs. 2%; p < 0.001) and diabetes (34% vs. 13%; p = 0.01) were more common in cases. Over 102 days (interquartile range: 62 to 214 days) of median follow-up, 16 (46%) developed MACE; 38% of MACE occurred with normal ejection fraction. There was a 4-fold increased risk of MACE with troponin T of ≥1.5 ng/ml (hazard ratio: 4.0; 95% confidence interval: 1.5 to 10.9; p = 0.003). Steroids were administered in 89%, and lower steroids doses were associated with higher residual troponin and higher MACE rates. CONCLUSIONS Myocarditis after ICI therapy may be more common than appreciated, occurs early after starting treatment, has a malignant course, and responds to higher steroid doses.
Collapse
Affiliation(s)
- Syed S Mahmood
- Cardiology Division, New York-Presbyterian Hospital, Weill Cornell Medical Center, New York, New York; Cardiac MR PET CT Program, Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts
| | - Michael G Fradley
- Cardio-Oncology Program, H. Lee Moffitt Cancer Center & Research Institute and University of South Florida Division of Cardiovascular Medicine, Tampa, Florida
| | - Justine V Cohen
- Division of Oncology and Hematology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Anju Nohria
- Cardio-Oncology Program, Division of Cardiology, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Kerry L Reynolds
- Division of Oncology and Hematology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Lucie M Heinzerling
- Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nurnberg (FAU), Germany
| | - Ryan J Sullivan
- Division of Oncology and Hematology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Rongras Damrongwatanasuk
- Cardio-Oncology Program, H. Lee Moffitt Cancer Center & Research Institute and University of South Florida Division of Cardiovascular Medicine, Tampa, Florida
| | - Carol L Chen
- Cardiology Division, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York, New York
| | - Dipti Gupta
- Cardiology Division, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York, New York
| | - Michael C Kirchberger
- Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nurnberg (FAU), Germany
| | - Magid Awadalla
- Cardiac MR PET CT Program, Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts
| | - Malek Z O Hassan
- Cardiac MR PET CT Program, Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts
| | - Javid J Moslehi
- Cardio-Oncology Program, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Sachin P Shah
- Cardiology Division, Lahey Hospital & Medical Center, Burlington, Massachusetts
| | - Sarju Ganatra
- Cardiology Division, Lahey Hospital & Medical Center, Burlington, Massachusetts
| | - Paaladinesh Thavendiranathan
- Ted Rogers Program in Cardiotoxicity Prevention, Peter Munk Cardiac Center, Division of Cardiology Toronto General Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Donald P Lawrence
- Division of Oncology and Hematology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - John D Groarke
- Cardio-Oncology Program, Division of Cardiology, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Tomas G Neilan
- Cardiac MR PET CT Program, Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts; Cardio-Oncology Program, Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts.
| |
Collapse
|
2691
|
Gorry C, McCullagh L, O'Donnell H, Barrett S, Schmitz S, Barry M, Curtin K, Beausang E, Barry R, Coyne I. Neoadjuvant treatment for malignant and metastatic cutaneous melanoma. THE COCHRANE DATABASE OF SYSTEMATIC REVIEWS 2018. [DOI: 10.1002/14651858.cd012974] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Claire Gorry
- National Centre for Pharmacoeconomics, St James's Hospital; St James's Hospital Dublin Ireland 8
| | - Laura McCullagh
- Trinity Centre for Health Sciences, St James's Hospital; Pharmacology and Therapeutics; St James's Hospital Dublin Ireland Dublin 8
| | - Helen O'Donnell
- Trinity Centre for Health Sciences, St James's Hospital; Pharmacology and Therapeutics; St James's Hospital Dublin Ireland Dublin 8
| | - Sarah Barrett
- Trinity Centre for Health Sciences, St James's Hospital; Discipline of Radiation Therapy, School of Medicine; Trinity Centre for Health Sciences, James's St Dublin Ireland 8
| | - Susanne Schmitz
- Trinity Centre for Health Sciences, St James's Hospital; Pharmacology and Therapeutics; St James's Hospital Dublin Ireland Dublin 8
- Luxembourg Institute of Health; Department of Population Health; 1A-B, rue Thomas Edison Strassen Luxembourg 1445
| | - Michael Barry
- Trinity Centre for Health Sciences, St James's Hospital; Pharmacology and Therapeutics; St James's Hospital Dublin Ireland Dublin 8
| | - Kay Curtin
- Melanoma Support Ireland; Dublin Ireland
| | - Eamon Beausang
- St James's Hospital; Plastic and Reconstructive Surgery; Dublin Ireland 8
| | - Rupert Barry
- St James's Hospital; Dermatology; James Street Dublin Ireland 8
| | - Imelda Coyne
- Trinity College Dublin; School of Nursing & Midwifery; 24 D'Olier St Dublin Ireland 2
| |
Collapse
|
2692
|
Dillman RO, Cornforth AN, Nistor GI, McClay EF, Amatruda TT, Depriest C. Randomized phase II trial of autologous dendritic cell vaccines versus autologous tumor cell vaccines in metastatic melanoma: 5-year follow up and additional analyses. J Immunother Cancer 2018; 6:19. [PMID: 29510745 PMCID: PMC5840808 DOI: 10.1186/s40425-018-0330-1] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 02/26/2018] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Despite improved survival following checkpoint inhibitors, there is still a potential role for anti-cancer therapeutic vaccines. Because of biological heterogeneity and neoantigens resulting from each patient's mutanome, autologous tumor may be the best source of tumor-associated antigens (TAA) for vaccines. Ex vivo loading of autologous dendritic cells with TAA may be associated with superior clinical outcome compared to injecting irradiated autologous tumor cells. We conducted a randomized phase II trial to compare autologous tumor cell vaccines (TCV) and autologous dendritic cell vaccines (DCV) loaded with autologous TAA. METHODS Short-term autologous tumor cell lines were established from metastatic tumor. Vaccines were admixed with 500 micrograms of GM-CSF and injected weekly for 3 weeks, then at weeks 8, 12,16, 20, and 24. The primary endpoint was overall survival. Secondary objectives were identification of adverse events, and results of delayed type hypersensitivity (DTH) reactions to intradermal tumor cell injections. RESULTS Forty-two patients were randomized. All were followed from randomization until death or for five years; none were lost to follow-up. DCV was associated with longer survival: median 43.4 versus 20.5 months (95% CI, 18.6 to > 60 versus 9.3 to 32.3 months) and a 70% reduction in the risk of death (hazard ratio = 0.304, p = 0.0053, 95% CI, 0.131 to 0.702). Tumor DTH reactions were neither prognostic nor predictive. The most common treatment-related adverse events were mild to moderate local injection site reactions and flu-like symptoms; but grade 2 treatment-related adverse events were more frequent with TCV. Serum marker analyses at week-0 and week-4 showed that serum markers were similar at baseline in each arm, but differed after vaccination. CONCLUSIONS This is the only human clinical trial comparing DCV and TCV as platforms for autologous TAA presentation. DCV was associated with minimal toxicity and long-term survival in patients with metastatic melanoma. DTH to autologous tumor cells was neither prognostic for survival nor predictive of benefit for either vaccine. TRIAL REGISTRATION Clinical trials.gov NCT00948480 retrospectively registered 28 July 2009.
Collapse
Affiliation(s)
- Robert O. Dillman
- Hoag Cancer Institute, Newport Beach, CA 92660 USA
- AIVITA Biomedical, Inc., Irvine, CA USA
| | | | | | - Edward F. McClay
- California Cancer Associates for Research and Excellence (cCARE), Institute for Melanoma Research & Education, Encinitas, CA USA
| | | | | |
Collapse
|
2693
|
Abstract
Immunotherapy has emerged as a major therapeutic modality in oncology. Currently, however, the majority of patients with cancer do not derive benefit from these treatments. Vascular abnormalities are a hallmark of most solid tumours and facilitate immune evasion. These abnormalities stem from elevated levels of proangiogenic factors, such as VEGF and angiopoietin 2 (ANG2); judicious use of drugs targeting these molecules can improve therapeutic responsiveness, partially owing to normalization of the abnormal tumour vasculature that can, in turn, increase the infiltration of immune effector cells into tumours and convert the intrinsically immunosuppressive tumour microenvironment (TME) to an immunosupportive one. Immunotherapy relies on the accumulation and activity of immune effector cells within the TME, and immune responses and vascular normalization seem to be reciprocally regulated. Thus, combining antiangiogenic therapies and immunotherapies might increase the effectiveness of immunotherapy and diminish the risk of immune-related adverse effects. In this Perspective, we outline the roles of VEGF and ANG2 in tumour immune evasion and progression, and discuss the evidence indicating that antiangiogenic agents can normalize the TME. We also suggest ways that antiangiogenic agents can be combined with immune-checkpoint inhibitors to potentially improve patient outcomes, and highlight avenues of future research.
Collapse
|
2694
|
Abstract
INTRODUCTION Urothelial bladder cancer is one of the most predominant malignancies worldwide with a poor prognosis when presented at an advanced or metastatic stage. Improving the therapeutic landscape in this setting has been an unmet medical need. Palliative cisplatin-based chemotherapy is currently the standard of care in first line therapies, but many patients are ineligible and few alternative therapies exist. Moreover second-line chemotherapy has minimal activity. Recently, immune-checkpoint inhibitors have shifted the therapeutic armamentarium of bladder cancer and it is now necessary to redesign the therapeutic paradigm. Areas covered: In this article, we focus on the development of durvalumab and provide an overview of the safety, activity, efficacy and future perspectives of this drug in urothelial carcinoma. Expert commentary: Durvalumab is a well-tolerated drug and demonstrated major and durable response in advanced bladder cancer. Combinations with durvalumab will probably emerge as promising therapeutic strategies for the treatment of urothelial carcinoma. Further research efforts are needed to identify predictive biomarkers of response to immune-oncology agents.
Collapse
Affiliation(s)
- Pernelle Lavaud
- a Gustave Roussy, Department de Medicine Oncologique & INSERM U981 , Université Paris-Saclay , Villejuif , France
| | - Zineb Hamilou
- a Gustave Roussy, Department de Medicine Oncologique & INSERM U981 , Université Paris-Saclay , Villejuif , France
| | - Yohann Loriot
- a Gustave Roussy, Department de Medicine Oncologique & INSERM U981 , Université Paris-Saclay , Villejuif , France
| | - Christophe Massard
- b Drug Development Department (DITEP), Gustave Roussy , Université Paris-Sud, Université Paris-Saclay , Villejuif , France
| |
Collapse
|
2695
|
Mullinax JE, Hall M, Prabhakaran S, Weber J, Khushalani N, Eroglu Z, Brohl AS, Markowitz J, Royster E, Richards A, Stark V, Zager JS, Kelley L, Cox C, Sondak VK, Mulé JJ, Pilon-Thomas S, Sarnaik AA. Combination of Ipilimumab and Adoptive Cell Therapy with Tumor-Infiltrating Lymphocytes for Patients with Metastatic Melanoma. Front Oncol 2018; 8:44. [PMID: 29552542 PMCID: PMC5840208 DOI: 10.3389/fonc.2018.00044] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 02/12/2018] [Indexed: 01/09/2023] Open
Abstract
PURPOSE Adoptive cell therapy (ACT) using tumor-infiltrating lymphocytes (TIL) for metastatic melanoma can be highly effective, but attrition due to progression before TIL administration (32% in prior institutional experience) remains a limitation. We hypothesized that combining ACT with cytotoxic T lymphocyte-associated antigen 4 blockade would decrease attrition and allow more patients to receive TIL. EXPERIMENTAL DESIGN Thirteen patients with metastatic melanoma were enrolled. Patients received four doses of ipilimumab (3 mg/kg) beginning 2 weeks prior to tumor resection for TIL generation, then 1 week after resection, and 2 and 5 weeks after preconditioning chemotherapy and TIL infusion followed by interleukin-2. The primary endpoint was safety and feasibility. Secondary endpoints included of clinical response at 12 weeks and at 1 year after TIL transfer, progression free survival (PFS), and overall survival (OS). RESULTS All patients received at least two doses of ipilimumab, and 12 of the 13 (92%) received TIL. A median of 6.5 × 1010 (2.3 × 1010 to 1.0 × 1011) TIL were infused. At 12 weeks following infusion, there were five patients who experienced objective response (38.5%), four of whom continued in objective response at 1 year and one of which became a complete response at 52 months. Median progression-free survival was 7.3 months (95% CI 6.1-29.9 months). Grade ≥ 3 immune-related adverse events included hypothyroidism (3), hepatitis (2), uveitis (1), and colitis (1). CONCLUSION Ipilimumab plus ACT for metastatic melanoma is feasible, well tolerated, and associated with a low rate of attrition due to progression during cell expansion. This combination approach serves as a model for future efforts to improve the efficacy of ACT.
Collapse
Affiliation(s)
- John E. Mullinax
- Sarcoma Department, Moffitt Cancer Center, Tampa, FL, United States
- Immunology Department, Moffitt Cancer Center, Tampa, FL, United States
| | - MacLean Hall
- Immunology Department, Moffitt Cancer Center, Tampa, FL, United States
| | | | - Jeffrey Weber
- NYU Langone Medical Center, New York, NY, United States
| | - Nikhil Khushalani
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, FL, United States
| | - Zeynep Eroglu
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, FL, United States
| | - Andrew S. Brohl
- Sarcoma Department, Moffitt Cancer Center, Tampa, FL, United States
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, FL, United States
| | - Joseph Markowitz
- Immunology Department, Moffitt Cancer Center, Tampa, FL, United States
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, FL, United States
| | - Erica Royster
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, FL, United States
| | - Allison Richards
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, FL, United States
| | - Valerie Stark
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, FL, United States
| | - Jonathan S. Zager
- Sarcoma Department, Moffitt Cancer Center, Tampa, FL, United States
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, FL, United States
| | - Linda Kelley
- Immunology Department, Moffitt Cancer Center, Tampa, FL, United States
| | - Cheryl Cox
- Immunology Department, Moffitt Cancer Center, Tampa, FL, United States
| | - Vernon K. Sondak
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, FL, United States
| | - James J. Mulé
- Immunology Department, Moffitt Cancer Center, Tampa, FL, United States
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, FL, United States
| | - Shari Pilon-Thomas
- Immunology Department, Moffitt Cancer Center, Tampa, FL, United States
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, FL, United States
| | - Amod A. Sarnaik
- Immunology Department, Moffitt Cancer Center, Tampa, FL, United States
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, FL, United States
| |
Collapse
|
2696
|
|
2697
|
Turajlic S, Gore M, Larkin J. First report of overall survival for ipilimumab plus nivolumab from the phase III Checkmate 067 study in advanced melanoma. Ann Oncol 2018; 29:542-543. [PMID: 29360923 DOI: 10.1093/annonc/mdy020] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- S Turajlic
- Melanoma Unit, The Royal Marsden NHS Foundation Trust, London, UK; Translational Cancer Therapeutics Laboratory, The Francis Crick Institute, London, UK
| | - M Gore
- Melanoma Unit, The Royal Marsden NHS Foundation Trust, London, UK
| | - J Larkin
- Melanoma Unit, The Royal Marsden NHS Foundation Trust, London, UK.
| |
Collapse
|
2698
|
Moskovitz JM, Ferris RL. Tumor Immunology and Immunotherapy for Head and Neck Squamous Cell Carcinoma. J Dent Res 2018; 97:622-626. [PMID: 29489423 DOI: 10.1177/0022034518759464] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The immune system plays an important role in the evolution of malignancy and has become an important target for novel antineoplastic agents. This review article focuses on key features of tumor immunology, including the role of immunotherapy in general and as it pertains to head and neck squamous cell carcinoma. Side effects, resistance mechanisms, and therapeutic monitoring strategies pertaining to immunotherapy are discussed.
Collapse
Affiliation(s)
- J M Moskovitz
- 1 Department of Otolaryngology, University of Pittsburgh, Pittsburgh, PA, USA
| | - R L Ferris
- 2 UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| |
Collapse
|
2699
|
Fujimura T, Kambayashi Y, Tanita K, Sato Y, Hidaka T, Otsuka A, Tanaka H, Furudate S, Hashimoto A, Aiba S. HLA-DRB1*04:05 in two cases of Vogt-Koyanagi-Harada disease-like uveitis developing from an advanced melanoma patient treated by sequential administration of nivolumab and dabrafenib/trametinib therapy. J Dermatol 2018; 45:735-737. [PMID: 29488243 DOI: 10.1111/1346-8138.14273] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 01/26/2018] [Indexed: 12/31/2022]
Abstract
Although uveitis is reported as a rare adverse event (AE) associated with dabrafenib/trametinib therapy or nivolumab, the occurrence of severe uveitis is extremely rare. We describe two cases of Vogt-Koyanagi-Harada (VKH)-like uveitis developing after the sequential administration of nivolumab and dabrafenib/trametinib therapy. Interestingly, both cases had HLA-DRB1*04:05, which is strongly associated with VKH disease, and achieved biologically complete remission after the treatment for uveitis. Our cases suggest a possible correlation between VKH-like uveitis as an AE and the clinical outcomes of sequential administration of nivolumab and dabrafenib/trametinib therapy for the treatment of advanced melanoma.
Collapse
Affiliation(s)
- 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
| | - Kayo Tanita
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yota Sato
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takanori Hidaka
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Astushi Otsuka
- Department of Dermatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | | | - Sadanori Furudate
- 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
|
2700
|
Marconcini R, Spagnolo F, Stucci LS, Ribero S, Marra E, Rosa FD, Picasso V, Di Guardo L, Cimminiello C, Cavalieri S, Orgiano L, Tanda E, Spano L, Falcone A, Queirolo P, for the Italian Melanoma Intergroup (IMI). Current status and perspectives in immunotherapy for metastatic melanoma. Oncotarget 2018; 9:12452-12470. [PMID: 29552325 PMCID: PMC5844761 DOI: 10.18632/oncotarget.23746] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2017] [Accepted: 11/03/2017] [Indexed: 12/31/2022] Open
Abstract
Metastatic melanoma was the first malignancy in which immune checkpoint inhibitors demonstrated their successful efficacy. Currently, the knowledge on the interaction between the immune system and malignant disease is steadily increasing and new drugs and therapeutic strategies are overlooking in the clinical scenario. To provide a comprehensive overview of immune modulating drugs currently available in the treatment of melanoma as well as to discuss of possible future strategies in the metastatic melanoma setting, the present review aims at analyzing controversial aspects about the optimal immunomodulating treatment sequences, the search for biomarkers of efficacy of immunocheckpoint inhibitors, and innovative combinations of drugs currently under investigation.
Collapse
Affiliation(s)
- Riccardo Marconcini
- Unit of Medical Oncology 2, Azienda Ospedaliera-Universitaria
Pisana, Department of Translational Research and New Technologies in Medicine and
Surgery, University of Pisa, Italy
| | - Francesco Spagnolo
- Department of Medical Oncology, IRCCS AOU San Martino-Istituto
Nazionale per la Ricerca sul Cancro, Genova, Italy
| | - Luigia Stefania Stucci
- Medical Oncology Unit, Department of Biomedical Sciences and
Clinical Oncology, University of Bari, Bari, Italy
| | - Simone Ribero
- Dermatologic Clinic, Department of Medical Sciences,
University of Turin, Turin, Italy
| | - Elena Marra
- Dermatologic Clinic, Department of Medical Sciences,
University of Turin, Turin, Italy
| | - Francesco De Rosa
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei
Tumori, IRST IRCCS, Meldola, Italy
| | - Virginia Picasso
- Department of Medical Oncology, IRCCS AOU San Martino-Istituto
Nazionale per la Ricerca sul Cancro, Genova, Italy
| | | | | | | | - Laura Orgiano
- AOU Cagliari, Department of Medical Oncology, University of
Cagliari, Cagliari, Italy
| | - Enrica Tanda
- Department of Medical Oncology, IRCCS AOU San Martino-Istituto
Nazionale per la Ricerca sul Cancro, Genova, Italy
| | - Laura Spano
- Department of Medical Oncology, IRCCS AOU San Martino-Istituto
Nazionale per la Ricerca sul Cancro, Genova, Italy
| | - Alfredo Falcone
- Unit of Medical Oncology 2, Azienda Ospedaliera-Universitaria
Pisana, Department of Translational Research and New Technologies in Medicine and
Surgery, University of Pisa, Italy
| | - Paola Queirolo
- Department of Medical Oncology, IRCCS AOU San Martino-Istituto
Nazionale per la Ricerca sul Cancro, Genova, Italy
| | - for the Italian Melanoma Intergroup (IMI)
- Unit of Medical Oncology 2, Azienda Ospedaliera-Universitaria
Pisana, Department of Translational Research and New Technologies in Medicine and
Surgery, University of Pisa, Italy
- Department of Medical Oncology, IRCCS AOU San Martino-Istituto
Nazionale per la Ricerca sul Cancro, Genova, Italy
- Medical Oncology Unit, Department of Biomedical Sciences and
Clinical Oncology, University of Bari, Bari, Italy
- Dermatologic Clinic, Department of Medical Sciences,
University of Turin, Turin, Italy
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei
Tumori, IRST IRCCS, Meldola, Italy
- Fondazione IRCCS Istituto Nazionale dei Tumori, Milan,
Italy
- AOU Cagliari, Department of Medical Oncology, University of
Cagliari, Cagliari, Italy
| |
Collapse
|