2701
|
Recent advances in therapeutic strategies for unresectable or metastatic melanoma and real-world data in Japan. Int J Clin Oncol 2018; 24:1508-1514. [PMID: 29470725 DOI: 10.1007/s10147-018-1246-y] [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: 01/10/2018] [Accepted: 01/17/2018] [Indexed: 12/13/2022]
Abstract
New therapeutic strategies including immunotherapy and selective molecular target inhibitors have brought about a new era in the treatment of patients with advanced melanoma. In Japan, the immune checkpoint inhibitors ipilimumab, nivolumab and pembrolizumab, the BRAF inhibitor (BRAFi) vemurafenib, dabrafenib and MEK inhibitor (MEKi) trametinib have been available for the treatment of unresectable and metastatic melanoma. The BRAFi + MEKi combination shows high response rates (60-70%) and rapid response induction associated with symptom control, with a progression-free survival of 12 months. Nivolumab and pembrolizumab offer moderate response rates (30-40%) and long survival (3- to 5-year survival: 30-50%). In Japan, treatment options for the first-line setting frequently include nivolumab or pembrolizumab monotherapy and BRAFi + MEKi combinations (for patients with BRAF-mutant melanoma). Ipilimumab is included in the second-line setting, and the nivolumab + ipilimumab combination has not been approved yet in Japan. Although these medications have demonstrated impressive efficacy, the clinical trials and real-world data have shown that the clinical benefit is not fully satisfactory. We have to carefully manage a new class of adverse events due to these medicines. Moreover, biomarkers are emerging with which we can identify a population that would experience more benefits without severe adverse events.
Collapse
|
2702
|
Rosner S, Kwong E, Shoushtari AN, Friedman CF, Betof AS, Brady MS, Coit DG, Callahan MK, Wolchok JD, Chapman PB, Panageas KS, Postow MA. Peripheral blood clinical laboratory variables associated with outcomes following combination nivolumab and ipilimumab immunotherapy in melanoma. Cancer Med 2018; 7:690-697. [PMID: 29468834 PMCID: PMC5852343 DOI: 10.1002/cam4.1356] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 11/20/2017] [Accepted: 12/28/2017] [Indexed: 01/27/2023] Open
Abstract
Both the combination of nivolumab + ipilimumab and single-agent anti-PD-1 immunotherapy have demonstrated survival benefit for patients with advanced melanoma. As the combination has a high rate of serious side effects, further analyses in randomized trials of combination versus anti-PD-1 immunotherapy are needed to understand who benefits most from the combination. Clinical laboratory values that were routinely collected in randomized studies may provide information on the relative benefit of combination immunotherapy. To prioritize which clinical laboratory factors to ultimately explore in these randomized studies, we performed a single-center, retrospective analysis of patients with advanced melanoma who received nivolumab + ipilimumab either as part of a clinical trial (n = 122) or commercial use (n = 87). Baseline routine laboratory values were correlated with overall survival (OS) and overall response rate (ORR). Kaplan-Meier estimation and Cox regression were performed. Median OS was 44.4 months, 95% CI (32.9, Not Reached). A total of 110 patients (53%) responded (CR/PR). Significant independent variables for favorable OS included the following: high relative eosinophils, high relative basophils, low absolute monocytes, low LDH, and a low neutrophil-to-lymphocyte ratio. These newly identified factors, along with those previously reported to be associated with anti-PD-1 monotherapy outcomes, should be studied in the randomized trials of nivolumab + ipilimumab versus anti-PD-1 monotherapies to determine whether they help define the patients who benefit most from the combination versus anti-PD-1 alone.
Collapse
Affiliation(s)
- Samuel Rosner
- Department of Medicine, Johns Hopkins Bayview Medical Center, Baltimore, Maryland
| | - Erica Kwong
- City University of New York at Hunter College, New York City, New York
| | - Alexander N Shoushtari
- Memorial Sloan Kettering Cancer Center, New York City, New York.,Weill Cornell Medical College, New York City, New York
| | - Claire F Friedman
- Memorial Sloan Kettering Cancer Center, New York City, New York.,Weill Cornell Medical College, New York City, New York
| | - Allison S Betof
- Memorial Sloan Kettering Cancer Center, New York City, New York
| | - Mary Sue Brady
- Memorial Sloan Kettering Cancer Center, New York City, New York
| | - Daniel G Coit
- Memorial Sloan Kettering Cancer Center, New York City, New York
| | - Margaret K Callahan
- Memorial Sloan Kettering Cancer Center, New York City, New York.,Weill Cornell Medical College, New York City, New York
| | - Jedd D Wolchok
- Memorial Sloan Kettering Cancer Center, New York City, New York.,Weill Cornell Medical College, New York City, New York.,Ludwig Center for Cancer Immunotherapy, New York City, New York
| | - Paul B Chapman
- Memorial Sloan Kettering Cancer Center, New York City, New York.,Weill Cornell Medical College, New York City, New York
| | | | - Michael A Postow
- Memorial Sloan Kettering Cancer Center, New York City, New York.,Weill Cornell Medical College, New York City, New York
| |
Collapse
|
2703
|
Yang J, Manson DK, Marr BP, Carvajal RD. Treatment of uveal melanoma: where are we now? Ther Adv Med Oncol 2018; 10:1758834018757175. [PMID: 29497459 PMCID: PMC5824910 DOI: 10.1177/1758834018757175] [Citation(s) in RCA: 210] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 01/12/2018] [Indexed: 12/15/2022] Open
Abstract
Uveal melanoma, a rare subset of melanoma, is the most common primary intraocular malignancy in adults. Despite effective primary therapy, nearly 50% of patients will develop metastatic disease. Outcomes for those with metastatic disease remain dismal due to a lack of effective therapies. The unique biology and immunology of uveal melanoma necessitates the development of dedicated management and treatment approaches. Ongoing efforts seek to optimize the efficacy of targeted therapy and immunotherapy in both the adjuvant and metastatic setting. This review provides a comprehensive, updated overview of disease biology and risk stratification, the management of primary disease, options for adjuvant therapy, and the current status of treatment strategies for metastatic disease.
Collapse
Affiliation(s)
- Jessica Yang
- Division of Hematology/Oncology, Columbia University Medical Center, New York, NY, USA
| | - Daniel K. Manson
- Division of Hematology/Oncology, Columbia University Medical Center, New York, NY, USA
| | - Brian P. Marr
- Department of Ophthalmology, Columbia University Medical Center, New York, NY, USA
| | - Richard D. Carvajal
- Assistant Professor of Medicine, Director of Experimental Therapeutics and Melanoma Services, Division of Hematology/Oncology, Columbia University Medical Center, 177 Fort Washington Avenue, MHB 6GN-435, New York, NY 10032, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY, USA
| |
Collapse
|
2704
|
Bello DM, Ariyan CE. Adjuvant Therapy in the Treatment of Melanoma. Ann Surg Oncol 2018; 25:1807-1813. [PMID: 29468608 DOI: 10.1245/s10434-018-6376-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Indexed: 12/26/2022]
Affiliation(s)
- Danielle M Bello
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Charlotte E Ariyan
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA.
| |
Collapse
|
2705
|
The Role of the Estrogen Pathway in the Tumor Microenvironment. Int J Mol Sci 2018; 19:ijms19020611. [PMID: 29463044 PMCID: PMC5855833 DOI: 10.3390/ijms19020611] [Citation(s) in RCA: 136] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 02/13/2018] [Accepted: 02/16/2018] [Indexed: 12/15/2022] Open
Abstract
Estrogen receptors are broadly expressed in many cell types involved in the innate and adaptive immune responses, and differentially regulate the production of cytokines. While both genomic and non-genomic tumor cell promoting mechanisms of estrogen signaling are well characterized in multiple carcinomas including breast, ovarian, and lung, recent investigations have identified a potential immune regulatory role of estrogens in the tumor microenvironment. Tumor immune tolerance is a well-established mediator of oncogenesis, with increasing evidence indicating the importance of the immune response in tumor progression. Immune-based therapies such as antibodies that block checkpoint signals have emerged as exciting therapeutic approaches for cancer treatment, offering durable remissions and prolonged survival. However, only a subset of patients demonstrate clinical response to these agents, prompting efforts to elucidate additional immunosuppressive mechanisms within the tumor microenvironment. Evidence drawn from multiple cancer types, including carcinomas traditionally classified as non-immunogenic, implicate estrogen as a potential mediator of immunosuppression through modulation of protumor responses independent of direct activity on tumor cells. Herein, we review the interplay between estrogen and the tumor microenvironment and the clinical implications of endocrine therapy as a novel treatment strategy within immuno-oncology.
Collapse
|
2706
|
|
2707
|
Ahern E, Harjunpää H, O'Donnell JS, Allen S, Dougall WC, Teng MWL, Smyth MJ. RANKL blockade improves efficacy of PD1-PD-L1 blockade or dual PD1-PD-L1 and CTLA4 blockade in mouse models of cancer. Oncoimmunology 2018; 7:e1431088. [PMID: 29872559 DOI: 10.1080/2162402x.2018.1431088] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 01/15/2018] [Indexed: 01/05/2023] Open
Abstract
Receptor activator of NF-κB ligand (RANKL) and its receptor RANK, are members of the tumor necrosis factor and receptor superfamilies, respectively. Antibodies targeting RANKL have recently been evaluated in combination with anti-CTLA4 in case reports of human melanoma and mouse models of cancer. However, the efficacy of anti-RANKL in combination with antibodies targeting other immune checkpoint receptors such as PD1 has not been reported. In this study, we demonstrated that blockade of RANKL improves anti-metastatic activity of antibodies targeting PD1/PD-L1 and improves subcutaneous growth suppression in mouse models of melanoma, prostate and colon cancer. Suppression of experimental lung metastasis following combination anti-RANKL with anti-PD1 requires NK cells and IFN-γ, whereas subcutaneous tumor growth suppression with this combination therapy is attenuated in the absence of T cells and IFN-γ. Furthermore, addition of anti-RANKL to anti-PD1 and anti-CTLA4 resulted in superior anti-tumor responses, irrespective of the ability of anti-CTLA4 isotype to engage activating FcR, and concurrent or delayed RANKL blockade was most effective. Early-during-treatment assessment reveals this triple combination therapy compared to dual anti-PD1 and anti-CTLA4 combination therapy further increased the proportion of tumor-infiltrating CD4+ and CD8+ T cells that can produce both IFN-γ and TNF. Finally, RANKL expression appears to identify tumor-specific CD8+ T cells expressing higher levels of PD1 which can be modulated by anti-PD1. These data set the scene for clinical evaluation of denosumab use in patients receiving contemporary immune checkpoint blockade.
Collapse
Affiliation(s)
- Elizabeth Ahern
- Department of Immunology, Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia.,Division of Cancer Care Services, Medical Oncology, Royal Brisbane and Women's Hospital, Herston, Queensland, Australia
| | - Heidi Harjunpää
- Department of Immunology, Cancer Immunoregulation and Immunotherapy Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia.,Faculty of Medicine, University of Queensland, Herston, Queensland, Australia
| | - Jake S O'Donnell
- Department of Immunology, Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia.,Department of Immunology, Cancer Immunoregulation and Immunotherapy Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia.,Faculty of Medicine, University of Queensland, Herston, Queensland, Australia
| | - Stacey Allen
- Department of Immunology, Cancer Immunoregulation and Immunotherapy Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - William C Dougall
- Department of Immunology, Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia.,Department of Immunology, Immuno-oncology Discovery Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Michele W L Teng
- Department of Immunology, Cancer Immunoregulation and Immunotherapy Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia.,Faculty of Medicine, University of Queensland, Herston, Queensland, Australia
| | - Mark J Smyth
- Department of Immunology, Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia.,Faculty of Medicine, University of Queensland, Herston, Queensland, Australia
| |
Collapse
|
2708
|
|
2709
|
[Current aspects in the prognosis of advanced melanoma]. Hautarzt 2018; 69:249-259. [PMID: 29396638 DOI: 10.1007/s00105-018-4124-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The therapy of metastatic melanoma has changed rapidly in recent years. Immune checkpoint blockade and targeted therapy have replaced less effective chemotherapies. New clinical studies also point towards a substantial benefit of these drugs for the adjuvant treatment of high-risk patients. Thus, the prognosis of advanced melanoma has improved. Nevertheless, it remains a life-threatening condition due to frequent relapses and progression of the disease. This article aims at providing an overview of current treatment strategies for metastasized melanoma and their impact on prognosis of the disease. In addition, changes in the recently published American Joint Committee of Cancer (AJCC) classification identifying groups at risk will be highlighted.
Collapse
|
2710
|
Affiliation(s)
- Pia Kvistborg
- Division of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Amsterdam, Netherlands.
| | - Jonathan W Yewdell
- Cellular Biology Section, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, Bethesda, MD 20892, USA
| |
Collapse
|
2711
|
Affiliation(s)
| | - Justin F Gainor
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
2712
|
Audibert C, Stuntz M, Glass D. Treatment Sequencing in Advanced BRAF-Mutant Melanoma Patients: Current Practice in the United States. J Pharm Technol 2018; 34:17-23. [PMID: 34860980 DOI: 10.1177/8755122517747089] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Background: Treatment of advanced BRAF-mutant melanoma has changed dramatically in the past 3 years thanks to the approval of new immunotherapy and targeted therapy agents. Objectives: The goal of our survey was to investigate when immunotherapy and targeted therapy are used in the management of advanced melanoma patients and whether differences exist between the types of setting. Methods: Oncologists from academic centers, community-based centers, and private clinics were invited to participate in an online survey. Survey questions addressed the proportion of BRAF-mutant patients per treatment line, proportion of patients on targeted therapy and immunotherapy available in the United States, and reasons for prescribing each drug class. Results: A total of 101 physicians completed the survey, of which 47 worked in a private clinic, 33 in an academic center, and 21 in a community-based center. Academic center participants tended to see more severe patients (P < .001) and had more patients in second-line treatment than participants from other setting types. In addition, academic center physicians had more patients in clinical trials (P < .001), and they prescribed the ipilimumab and nivolumab combination more frequently. In terms of sequencing, all participants used targeted therapy for severe or rapidly progressing patients and immunotherapy for those who were less severe or slowly progressing. Conclusions: The findings illustrate the differences in treatment approach per type of setting, with patients in academic centers more likely to receive recently approved products or to be enrolled in clinical trials than those in community-based settings.
Collapse
|
2713
|
Khushalani NI. Duration of Anti-Programmed Death-1 Therapy in Advanced Melanoma: How Much of a Good Thing Is Enough? J Clin Oncol 2018; 36:1649-1653. [PMID: 29389234 DOI: 10.1200/jco.2017.76.8275] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The Oncology Grand Rounds series is designed to place original reports published in the Journal into clinical context. A case presentation is followed by a description of diagnostic and management challenges, a review of the relevant literature, and a summary of the authors' suggested management approaches. The goal of this series is to help readers better understand how to apply the results of key studies, including those published in Journal of Clinical Oncology, to patients seen in their own clinical practice. A 53-year-old healthy man presented with recurrent in-transit melanoma of the right lower extremity. Eight years prior he had undergone wide local excision and sentinel lymph node biopsy for invasive melanoma of the anteromedial aspect of the distal right thigh. Pathology revealed an ulcerated melanoma, Breslow depth 3.5 mm, and with one involved micrometastatic inguinal lymph node. Staging studies did not demonstrate distant metastases. Superficial inguinal node dissection was performed and did not identify any additional metastatic nodes of 14 retrieved for a final pathologic staging of T3bN1aM0 (stage IIIB) cutaneous melanoma. He received 12 months of adjuvant high-dose interferon alfa-2b. Two years later, he developed a 1.2-cm subcutaneous focus of in-transit recurrence approximately 4 cm proximal to the original melanoma site in the right thigh, which was treated with surgical resection followed by adjuvant radiotherapy. Over the next 4 years, he underwent six additional surgeries for isolated in-transit recurrences affecting the same limb. He was referred for therapeutic options at the time of his latest in-transit recurrence. Examination revealed three palpable subcutaneous nodules in the right thigh in the setting of lymphedema. A core biopsy confirmed recurrent melanoma (Fig 1). Whole-body fluorodeoxyglucose positron emission tomography imaging revealed at least 17 hypermetabolic cutaneous and subcutaneous nodules in the right thigh, four fluorodeoxyglucose-avid nodules below the right knee, but no distant metastases (Fig 2A). Brain magnetic resonance imaging was normal. His serum chemistry profile, including lactate dehydrogenase, was normal. Molecular analysis demonstrated presence of BRAF V600E mutation in the tumor. After multidisciplinary evaluation, an isolated limb infusion procedure of the right lower extremity was not believed to be feasible, secondary to the proximal extent of the recurrence. Therapy was initiated with pembrolizumab at 2 mg/kg intravenously every 3 weeks.
Collapse
|
2714
|
Haag GM, Zoernig I, Hassel JC, Halama N, Dick J, Lang N, Podola L, Funk J, Ziegelmeier C, Juenger S, Bucur M, Umansky L, Falk CS, Freitag A, Karapanagiotou-Schenkel I, Beckhove P, Enk A, Jaeger D. Phase II trial of ipilimumab in melanoma patients with preexisting humoural immune response to NY-ESO-1. Eur J Cancer 2018; 90:122-129. [PMID: 29306769 DOI: 10.1016/j.ejca.2017.12.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Accepted: 12/07/2017] [Indexed: 11/20/2022]
Abstract
BACKGROUND Immune checkpoint therapy has dramatically changed treatment options in patients with metastatic melanoma. However, a relevant part of patients still does not respond to treatment. Data regarding the prognostic or predictive significance of preexisting immune responses against tumour antigens are conflicting. Retrospective data suggested a higher clinical benefit of ipilimumab in melanoma patients with preexisting NY-ESO-1-specific immunity. PATIENTS AND METHODS Twenty-five patients with previously untreated or treated metastatic melanoma and preexisting humoural immune response against NY-ESO-1 received ipilimumab at a dose of 10 mg/kg in week 1, 4, 7, 10 followed by 3-month maintenance treatment for a maximum of 48 weeks. Primary endpoint was the disease control rate (irCR, irPR or irSD) according to immune-related response criteria (irRC). Secondary endpoints included the disease control rate according to RECIST criteria, progression-free survival and overall survival (OS). Humoural and cellular immune responses against NY-ESO-1 were analysed from blood samples. RESULTS Disease control rate according to irRC was 52%, irPR was observed in 36% of patients. Progression-free survival according to irRC was 7.8 months, according to RECIST criteria it was 2.9 months. Median OS was 22.7 months; the corresponding 1-year survival rate was 66.8%. Treatment-related grade 3 AEs occurred in 36% with no grade 4-5 AEs. No clear association was found between the presence of NY-ESO-1-specific cellular or humoural immune responses and clinical activity. CONCLUSION Ipilimumab demonstrated clinically relevant activity within this biomarker-defined population. NY-ESO-1 positivity, as a surrogate for a preexisting immune response against tumour antigens, might help identifying patients with a superior outcome from immune checkpoint blockade. CLINICAL TRIAL INFORMATION NCT01216696.
Collapse
Affiliation(s)
- G M Haag
- Department of Medical Oncology, National Center for Tumor Diseases, University Hospital Heidelberg, Germany.
| | - I Zoernig
- Department of Medical Oncology, National Center for Tumor Diseases, University Hospital Heidelberg, Germany
| | - J C Hassel
- Department of Dermatology and National Center for Tumor Diseases, University Hospital Heidelberg, Germany
| | - N Halama
- Department of Medical Oncology, National Center for Tumor Diseases, University Hospital Heidelberg, Germany
| | - J Dick
- Department of Dermatology and National Center for Tumor Diseases, University Hospital Heidelberg, Germany
| | - N Lang
- Department of Dermatology and National Center for Tumor Diseases, University Hospital Heidelberg, Germany
| | - L Podola
- Translational Immunology, National Center for Tumor Diseases, Heidelberg, Germany
| | - J Funk
- Department of Medical Oncology, National Center for Tumor Diseases, University Hospital Heidelberg, Germany
| | - C Ziegelmeier
- Department of Medical Oncology, National Center for Tumor Diseases, University Hospital Heidelberg, Germany
| | - S Juenger
- Translational Immunology, National Center for Tumor Diseases, Heidelberg, Germany
| | - M Bucur
- Translational Immunology, National Center for Tumor Diseases, Heidelberg, Germany
| | - L Umansky
- Translational Immunology, National Center for Tumor Diseases, Heidelberg, Germany
| | - C S Falk
- Institute of Transplant Immunology, IFB-Tx, Hannover Medical School, Hannover, Germany
| | - A Freitag
- NCT Trial Center, National Center for Tumor Diseases, Heidelberg, Germany
| | | | - P Beckhove
- Translational Immunology, National Center for Tumor Diseases, Heidelberg, Germany; Regensburg Center for Interventional Immunology, University Hospital Regensburg, Germany
| | - A Enk
- Department of Dermatology and National Center for Tumor Diseases, University Hospital Heidelberg, Germany
| | - D Jaeger
- Department of Medical Oncology, National Center for Tumor Diseases, University Hospital Heidelberg, Germany; Clinical Cooperation Unit "Applied Tumor-Immunity", German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
2715
|
Wilson RAM, Evans TRJ, Fraser AR, Nibbs RJB. Immune checkpoint inhibitors: new strategies to checkmate cancer. Clin Exp Immunol 2018; 191:133-148. [PMID: 29139554 PMCID: PMC5758374 DOI: 10.1111/cei.13081] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 11/13/2017] [Accepted: 11/13/2017] [Indexed: 12/26/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) targeting cytotoxic T lymphocyte-associated protein-4 (CTLA-4) or programmed cell death protein 1 (PD-1) receptors have demonstrated remarkable efficacy in subsets of patients with malignant disease. This emerging treatment modality holds great promise for future cancer treatment and has engaged pharmaceutical research interests in tumour immunology. While ICIs can induce rapid and durable responses in some patients, identifying predictive factors for effective clinical responses has proved challenging. This review summarizes the mechanisms of action of ICIs and outlines important preclinical work that contributed to their development. We explore clinical data that has led to disease-specific drug licensing, and highlight key clinical trials that have revealed ICI efficacy across a range of malignancies. We describe how ICIs have been used as part of combination therapies, and explore their future prospects in this area. We conclude by discussing the incorporation of these new immunotherapeutics into precision approaches to cancer therapy.
Collapse
Affiliation(s)
- R. A. M. Wilson
- Institute of Infection, Immunity and Inflammation, University of GlasgowGlasgowUK
| | - T. R. J. Evans
- Cancer Research UK Beatson InstituteGlasgowUK
- Institute of Cancer Sciences, University of GlasgowGlasgowUK
| | - A. R. Fraser
- Institute of Infection, Immunity and Inflammation, University of GlasgowGlasgowUK
- Advanced TherapeuticsScottish National Blood Transfusion ServiceEdinburghUK
| | - R. J. B. Nibbs
- Institute of Infection, Immunity and Inflammation, University of GlasgowGlasgowUK
| |
Collapse
|
2716
|
Winer A, Bodor JN, Borghaei H. Identifying and managing the adverse effects of immune checkpoint blockade. J Thorac Dis 2018; 10:S480-S489. [PMID: 29593893 DOI: 10.21037/jtd.2018.01.111] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Immunotherapy has revolutionized the field of oncology. By inhibiting the cytotoxic T-lymphocyte-associated protein (CTLA-4) and programmed death-1 (PD-1) immune checkpoint pathways, multiple studies have demonstrated greatly improved survival in locally advanced and metastatic cancers including melanoma, renal, lung, gastric, and hepatocellular carcinoma. Trials in other malignancies are ongoing, and undoubtedly the number of drugs in this space will grow beyond the six currently approved by the Food and Drug Administration. However, by altering the immune response to fight cancer, a new class of side effects has emerged known as immune-related adverse events (irAEs). These adverse events are due to overactivation of the immune system in almost any organ of the body, and can occur at any point along a patient's treatment course. irAEs such as endocrinopathies (thyroiditis), colitis, and pneumonitis may occur more commonly. However, other organs such as the liver, heart, or brain may also be affected by immune overactivation and any of these side effects may become life threatening. This review presents an approach to promptly recognize and manage these toxicities, to hopefully minimize morbidity and mortality from irAEs.
Collapse
|
2717
|
McTyre E, Ayala-Peacock D, Contessa J, Corso C, Chiang V, Chung C, Fiveash J, Ahluwalia M, Kotecha R, Chao S, Attia A, Henson A, Hepel J, Braunstein S, Chan M. Multi-institutional competing risks analysis of distant brain failure and salvage patterns after upfront radiosurgery without whole brain radiotherapy for brain metastasis. Ann Oncol 2018; 29:497-503. [PMID: 29161348 DOI: 10.1093/annonc/mdx740] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND In this study, we use a competing risks analysis to assess factors predictive of early-salvage whole brain radiotherapy (WBRT) and early death after upfront stereotactic radiosurgery (SRS) alone for brain metastases in an attempt to identify populations that benefit less from upfront SRS. PATIENTS AND METHODS Patients from eight academic centers were treated with SRS for brain metastasis. Competing risks analysis was carried out for distant brain failure (DBF) versus death prior to DBF as well as for salvage SRS versus salvage WBRT versus death prior to salvage. Linear regression was used to determine predictors of the number of brain metastases at initial DBF (nDBF). RESULTS A total of 2657 patients were treated with upfront SRS alone. Multivariate analysis (MVA) identified an increased hazard of DBF associated with increasing number of brain metastases (P < 0.001), lowest SRS dose received (P < 0.001), and melanoma histology (P < 0.001), while there was a decreased hazard of DBF associated with increasing age (P < 0.001), KPS < 70 (P < 0.001), and progressive systemic disease (P = 0.004). MVA for first salvage SRS versus WBRT versus death prior to salvage revealed an increased hazard of first salvage WBRT seen with increasing number of brain metastases (P < 0.001) and a decreased hazard with widespread systemic disease (P = 0.002) and increasing age (P < 0.001). Variables associated with nDBF included age (P = 0.02), systemic disease status (P = 0.03), melanoma histology (P = 0.05), and initial number of brain metastases (P < 0.001). CONCLUSIONS Patients with a higher initial number of brain metastases were more likely to experience DBF, have a higher nDBF, and receive early-salvage WBRT, while patients who were older, had lower KPS, or had more systemic disease were more likely to experience death prior to DBF or salvage WBRT.
Collapse
Affiliation(s)
- E McTyre
- Department of Radiation Oncology, Wake Forest School of Medicine, Winston-Salem, USA.
| | - D Ayala-Peacock
- Department of Radiation Oncology, Wake Forest School of Medicine, Winston-Salem, USA; Department of Radiation Oncology, Vanderbilt University School of Medicine, Nashville, USA
| | - J Contessa
- Department of Therapeutic Radiology/Southeast Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, USA
| | - C Corso
- Department of Therapeutic Radiology/Southeast Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, USA
| | - V Chiang
- Department of Therapeutic Radiology/Southeast Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, USA; Department of Neurosurgery, Yale University School of Medicine, New Haven, USA
| | - C Chung
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, USA; Department of Radiation Oncology, Princess Margaret Cancer Center, Toronto, Canada, USA
| | - J Fiveash
- Department of Radiation Oncology, University of Alabama-Birmingham, Birmingham, USA
| | - M Ahluwalia
- Brain Tumor and Neuro-Oncology Center, Neurological Institute, Cleveland Clinic Foundation, Cleveland, USA
| | - R Kotecha
- Brain Tumor and Neuro-Oncology Center, Neurological Institute, Cleveland Clinic Foundation, Cleveland, USA
| | - S Chao
- Brain Tumor and Neuro-Oncology Center, Neurological Institute, Cleveland Clinic Foundation, Cleveland, USA
| | - A Attia
- Department of Radiation Oncology, Vanderbilt University School of Medicine, Nashville, USA
| | - A Henson
- Department of Radiation Oncology, Wake Forest School of Medicine, Winston-Salem, USA
| | - J Hepel
- Department of Radiation Oncology, Brown University Alpert Medical School, Providence, USA
| | - S Braunstein
- Department of Radiation Oncology, University of California San Francisco, San Francisco, USA
| | - M Chan
- Department of Radiation Oncology, Wake Forest School of Medicine, Winston-Salem, USA
| |
Collapse
|
2718
|
Teo MY, Rosenberg JE. Nivolumab for the treatment of urothelial cancers. Expert Rev Anticancer Ther 2018; 18:215-221. [DOI: 10.1080/14737140.2018.1432357] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Affiliation(s)
- Min Yuen Teo
- Department of Medicine, Division of Solid Tumor Oncology, Genitourinary Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jonathan E. Rosenberg
- Department of Medicine, Division of Solid Tumor Oncology, Genitourinary Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
2719
|
Hashimoto M, Kamphorst AO, Im SJ, Kissick HT, Pillai RN, Ramalingam SS, Araki K, Ahmed R. CD8 T Cell Exhaustion in Chronic Infection and Cancer: Opportunities for Interventions. Annu Rev Med 2018; 69:301-318. [PMID: 29414259 DOI: 10.1146/annurev-med-012017-043208] [Citation(s) in RCA: 456] [Impact Index Per Article: 65.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Antigen-specific CD8 T cells are central to the control of chronic infections and cancer, but persistent antigen stimulation results in T cell exhaustion. Exhausted CD8 T cells have decreased effector function and proliferative capacity, partly caused by overexpression of inhibitory receptors such as programmed cell death (PD)-1. Blockade of the PD-1 pathway has opened a new therapeutic avenue for reinvigorating T cell responses, with positive outcomes especially for patients with cancer. Other strategies to restore function in exhausted CD8 T cells are currently under evaluation-many in combination with PD-1-targeted therapy. Exhausted CD8 T cells comprise heterogeneous cell populations with unique differentiation and functional states. A subset of stem cell-like PD-1+ CD8 T cells responsible for the proliferative burst after PD-1 therapy has been recently described. A greater understanding of T cell exhaustion is imperative to establish rational immunotherapeutic interventions.
Collapse
Affiliation(s)
- Masao Hashimoto
- Emory Vaccine Center and Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia 30322, USA; , , , ,
| | - Alice O Kamphorst
- Emory Vaccine Center and Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia 30322, USA; , , , ,
| | - Se Jin Im
- Emory Vaccine Center and Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia 30322, USA; , , , ,
| | - Haydn T Kissick
- Emory Vaccine Center and Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia 30322, USA; , , , ,
- Department of Urology, Emory University School of Medicine, Atlanta, Georgia 30322, USA;
| | - Rathi N Pillai
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia 30322, USA; ,
| | - Suresh S Ramalingam
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia 30322, USA; ,
| | - Koichi Araki
- Emory Vaccine Center and Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia 30322, USA; , , , ,
| | - Rafi Ahmed
- Emory Vaccine Center and Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia 30322, USA; , , , ,
| |
Collapse
|
2720
|
Atkins MB, Hodi FS, Thompson JA, McDermott DF, Hwu WJ, Lawrence DP, Dawson NA, Wong DJ, Bhatia S, James M, Jain L, Robey S, Shu X, Homet Moreno B, Perini RF, Choueiri TK, Ribas A. Pembrolizumab Plus Pegylated Interferon alfa-2b or Ipilimumab for Advanced Melanoma or Renal Cell Carcinoma: Dose-Finding Results from the Phase Ib KEYNOTE-029 Study. Clin Cancer Res 2018; 24:1805-1815. [PMID: 29358500 DOI: 10.1158/1078-0432.ccr-17-3436] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 01/02/2018] [Accepted: 01/16/2018] [Indexed: 11/16/2022]
Abstract
Purpose: Pembrolizumab monotherapy, ipilimumab monotherapy, and pegylated interferon alfa-2b (PEG-IFN) monotherapy are active against melanoma and renal cell carcinoma (RCC). We explored the safety and preliminary antitumor activity of pembrolizumab combined with either ipilimumab or PEG-IFN in patients with advanced melanoma or RCC.Experimental Design: The phase Ib KEYNOTE-029 study (ClinicalTrials.gov, NCT02089685) included independent pembrolizumab plus reduced-dose ipilimumab and pembrolizumab plus PEG-IFN cohorts. Pembrolizumab 2 mg/kg every 3 weeks (Q3W) plus 4 doses of ipilimumab 1 mg/kg Q3W was tolerable if ≤6 of 18 patients experienced a dose-limiting toxicity (DLT). The target DLT rate for pembrolizumab 2 mg/kg Q3W plus PEG-IFN was 30%, with a maximum of 14 patients per dose level. Response was assessed per RECIST v1.1 by central review.Results: The ipilimumab cohort enrolled 22 patients, including 19 evaluable for DLTs. Six patients experienced ≥1 DLT. Grade 3 to 4 treatment-related adverse events occurred in 13 (59%) patients. Responses occurred in 5 of 12 (42%) patients with melanoma and 3 of 10 (30%) patients with RCC. In the PEG-IFN cohort, DLTs occurred in 2 of 14 (14%) patients treated at dose level 1 (PEG-IFN 1 μg/kg/week) and 2 of 3 (67%) patients treated at dose level 2 (PEG-IFN 2 μg/kg/week). Grade 3 to 4 treatment-related adverse events occurred in 10 of 17 (59%) patients. Responses occurred in 1 of 5 (20%) patients with melanoma and 2 of 12 (17%) patients with RCC.Conclusions: Pembrolizumab 2 mg/kg Q3W plus ipilimumab 1 mg/kg Q3W was tolerable and provided promising antitumor activity in patients with advanced melanoma or RCC. The maximum tolerated dose of pembrolizumab plus PEG-IFN had limited antitumor activity in this population. Clin Cancer Res; 24(8); 1805-15. ©2018 AACR.
Collapse
Affiliation(s)
- Michael B Atkins
- Georgetown-Lombardi Comprehensive Cancer Center, Washington, DC.
| | | | | | | | - Wen-Jen Hwu
- The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Nancy A Dawson
- Georgetown-Lombardi Comprehensive Cancer Center, Washington, DC
| | - Deborah J Wong
- University of California, Los Angeles, Los Angeles, California
| | | | - Marihella James
- The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Seth Robey
- Merck & Co., Inc., Kenilworth, New Jersey
| | - Xinxin Shu
- Merck & Co., Inc., Kenilworth, New Jersey
| | | | | | | | - Antoni Ribas
- University of California, Los Angeles, Los Angeles, California
| |
Collapse
|
2721
|
Immune Toxicity with Checkpoint Inhibition for Metastatic Melanoma: Case Series and Clinical Management. J Skin Cancer 2018; 2018:9602540. [PMID: 29610684 PMCID: PMC5828308 DOI: 10.1155/2018/9602540] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 10/31/2017] [Accepted: 11/06/2017] [Indexed: 12/16/2022] Open
Abstract
Immune checkpoint inhibitors (anti-PD-1 and anti-CTLA-4 antibodies) are a standard of care for advanced melanoma. Novel toxicities comprise immune-related adverse events (irAE). With increasing use, irAE require recognition, practical management strategies, and multidisciplinary care. We retrospectively evaluated the incidence, kinetics, and management of irAE in 41 patients receiving anti-PD-1 antibody therapy (pembrolizumab) for advanced melanoma. 63% received prior anti-CTLA-4 antibody therapy (ipilimumab). IrAE occurred in 54%, most commonly dermatological (24%), rheumatological (22%), and thyroid dysfunction (12%). Thyroiditis was characterised by a brief asymptomatic hyperthyroid phase followed by a symptomatic hypothyroid phase requiring thyroxine replacement. Transplant rejection doses of methylprednisolone were necessary to manage refractory hepatotoxicity. A bullous pemphigoid-like skin reaction with refractory pruritus responded to corticosteroids and neuropathic analgesia. Disabling grade 3-4 oligoarthritis required sulfasalazine therapy in combination with steroids. The median interval between the last dose of anti-CTLA-4 antibody and the first dose of anti-PD-1 therapy was 2.0 months (range: 0.4 to 22.4). Toxicities may occur late; this requires vigilance and multidisciplinary management which may allow effective anticancer therapy to continue. Management algorithms for thyroiditis, hypophysitis, arthralgia/arthritis, colitis, steroid-refractory hepatitis, and skin toxicity are discussed.
Collapse
|
2722
|
Agarwal PK, Sternberg CN. Clinical Trials Corner. Bladder Cancer 2018; 4:133-136. [PMID: 29430515 PMCID: PMC5798534 DOI: 10.3233/blc-189028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Affiliation(s)
- Piyush K Agarwal
- Urologic Oncology Branch, National Cancer Institute, Bethesda, MD, USA
| | - Cora N Sternberg
- Department of Medical Oncology, San Camillo Forlanini Hospital, Rome, Italy
| |
Collapse
|
2723
|
Lim SY, Lee JH, Diefenbach RJ, Kefford RF, Rizos H. Liquid biomarkers in melanoma: detection and discovery. Mol Cancer 2018; 17:8. [PMID: 29343260 PMCID: PMC5772714 DOI: 10.1186/s12943-018-0757-5] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 01/02/2018] [Indexed: 12/18/2022] Open
Abstract
A vast array of tumor-derived genetic, proteomic and cellular components are constantly released into the circulation of cancer patients. These molecules including circulating tumor DNA and RNA, proteins, tumor and immune cells are emerging as convenient and accurate liquid biomarkers of cancer. Circulating cancer biomarkers provide invaluable information on cancer detection and diagnosis, prognosticate patient outcomes, and predict treatment response. In this era of effective molecular targeted treatments and immunotherapies, there is now an urgent need to implement use of these circulating biomarkers in the clinic to facilitate personalized therapy. In this review, we present recent findings in circulating melanoma biomarkers, examine the challenges and promise of evolving technologies used for liquid biomarker discovery, and discuss future directions and perspectives in melanoma biomarker research.
Collapse
Affiliation(s)
- Su Yin Lim
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia.,Melanoma Institute Australia, Sydney, NSW, Australia
| | - Jenny H Lee
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia.,Melanoma Institute Australia, Sydney, NSW, Australia
| | - Russell J Diefenbach
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia.,Melanoma Institute Australia, Sydney, NSW, Australia
| | - Richard F Kefford
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia.,Melanoma Institute Australia, Sydney, NSW, Australia.,Department of Medical Oncology, Crown Princess Mary Cancer Centre, Westmead and Blacktown Hospitals, Sydney, NSW, Australia
| | - Helen Rizos
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia. .,Melanoma Institute Australia, Sydney, NSW, Australia. .,Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, 2 Technology Place, Sydney, NSW, 2109, Australia.
| |
Collapse
|
2724
|
Abstract
The incidence of melanoma continues to rise worldwide. Prior to 2010, there had been no progress in the treatment of advanced melanoma in living memory. Since then, immunotherapy has become a standard of care in the treatment of advanced melanoma. Nivolumab is a fully human monoclonal antibody against PD-1, which is a negative regulatory checkpoint in the T cells. The clinical benefit of nivolumab as a single agent is well established, with response rates of ≥40%, durable responses and a favorable tolerability profile. The combination of nivolumab and ipilimumab has also become a standard of care and the role of nivolumab in the adjuvant setting for high-risk patients has been recently confirmed.
Collapse
Affiliation(s)
- Fabio Gomes
- The Christie NHS Foundation Trust, Manchester, UK
| | | | - Paul Lorigan
- The Christie NHS Foundation Trust, Manchester, UK
| |
Collapse
|
2725
|
Liudahl SM, Coussens LM. B cells as biomarkers: predicting immune checkpoint therapy adverse events. J Clin Invest 2018; 128:577-579. [PMID: 29309049 DOI: 10.1172/jci99036] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Immune checkpoint inhibitors are becoming a cornerstone of cancer immunotherapy as a result of their clinical success in relieving immune suppression and driving durable antitumor T cell responses in certain subsets of patients. Unfortunately, checkpoint inhibition is also associated with treatment-related toxicities that result in a myriad of side effects, ranging from mild and manageable to severe and debilitating. In this issue of the JCI, Das and colleagues report an association between early therapy-induced changes in circulating B cells and an increased risk of high-grade immune-related adverse events (IRAEs) in patients treated with checkpoint inhibitors that target cytotoxic T lymphocyte-associated antigen-4 (CTLA4) and programmed cell death protein 1 (PD1). These findings identify potential predictive biomarkers for high-grade IRAEs that may be leveraged to improve patient monitoring and may prompt new treatment strategies to prevent IRAEs.
Collapse
|
2726
|
The dynamics of medical care in skin cancers. Curr Opin Oncol 2018; 30:105-106. [PMID: 29303788 DOI: 10.1097/cco.0000000000000435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
2727
|
Fujimura T, Kambayashi Y, Sato Y, Tanita K, Furudate S, Tsukada A, Tono H, Hashimoto A, Aiba S. Successful Treatment of Nivolumab-Resistant Multiple In-Transit Melanomas with Ipilimumab and Topical Imiquimod. Case Rep Oncol 2018. [PMID: 29515401 PMCID: PMC5836202 DOI: 10.1159/000485612] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Simultaneous or sequential, planned administration of ipilimumab could significantly enhance the antitumor effects of nivolumab in advanced melanoma patients. On the other hand, the efficacy of ipilimumab for nivolumab-resistant advanced melanoma is extremely poor. Therefore, additional supportive therapy for anti-PD-1 antibody therapy-resistant advanced melanoma has been widely investigated. In this report, we describe a case of multiple in-transit melanomas developing in a nivolumab-resistant patient successfully treated with ipilimumab in combination with imiquimod. Our present case suggested a possible therapy for nivolumab-resistant multiple in-transit melanomas using ipilimumab in combination with topical imiquimod.
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
| | - Yota Sato
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kayo Tanita
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Sadanori Furudate
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Akira Tsukada
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hisayuki Tono
- 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
|
2728
|
Ferguson PM, Long GV, Scolyer RA, Thompson JF. Impact of genomics on the surgical management of melanoma. Br J Surg 2018; 105:e31-e47. [PMID: 29341162 DOI: 10.1002/bjs.10751] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 09/28/2017] [Accepted: 10/10/2017] [Indexed: 12/19/2022]
Abstract
BACKGROUND Although surgery for early-stage melanoma offers the best chance of cure, recent advances in molecular medicine have revolutionized the management of late-stage melanoma, leading to significant improvements in clinical outcomes. Research into the genomic drivers of disease and cancer immunology has not only ushered in a new era of targeted and immune-based therapies for patients with metastatic melanoma, but has also provided new tools for monitoring disease recurrence and selecting therapeutic strategies. These advances present new opportunities and challenges to the surgeon treating patients with melanoma. METHODS The literature was reviewed to evaluate diagnostic and therapeutic advances in the management of cutaneous melanoma, and to highlight the impact of these advances on surgical decision-making. RESULTS Genomic testing is not required in the surgical management of primary melanoma, although it can provide useful information in some situations. Circulating nucleic acids from melanoma cells can be detected in peripheral blood to predict disease recurrence before it manifests clinically, but validation is required before routine clinical application. BRAF mutation testing is the standard of care for all patients with advanced disease to guide therapy, including the planning of surgery in adjuvant and neoadjuvant settings. CONCLUSION Surgery remains central for managing primary melanoma, and is an important element of integrated multidisciplinary care in advanced disease, particularly for patients with resectable metastases. The field will undergo further change as clinical trials address the relationships between surgery, radiotherapy and systemic therapy for patients with high-risk, early-stage and advanced melanoma.
Collapse
Affiliation(s)
- P M Ferguson
- Melanoma Institute Australia, Sydney, New South Wales, Australia
- Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia
- Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| | - G V Long
- Melanoma Institute Australia, Sydney, New South Wales, Australia
- Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia
- Department of Medical Oncology, Royal North Shore Hospital, Sydney, New South Wales, Australia
| | - R A Scolyer
- Melanoma Institute Australia, Sydney, New South Wales, Australia
- Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia
- Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| | - J F Thompson
- Melanoma Institute Australia, Sydney, New South Wales, Australia
- Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia
- Department of Melanoma and Surgical Oncology, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| |
Collapse
|
2729
|
Borrie AE, Maleki Vareki S. T Lymphocyte–Based Cancer Immunotherapeutics. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2018; 341:201-276. [DOI: 10.1016/bs.ircmb.2018.05.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
2730
|
Jain A, Shannon VR, Sheshadri A. Immune-Related Adverse Events: Pneumonitis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 995:131-149. [PMID: 30539509 DOI: 10.1007/978-3-030-02505-2_6] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Checkpoint inhibitors are part of the family of immunotherapies and are increasingly being used in a wide variety of cancers. Immune-related adverse events pose a major challenge in the treatment of cancer patients. Pneumonitis is a rare immune-related adverse event that presents in distinct patterns. The goal of this chapter is to instruct readers on the incidence and clinical manifestations of pneumonitis and to offer guidance in the evaluation and treatment of patients with pneumonitis.
Collapse
Affiliation(s)
- Akash Jain
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Vickie R Shannon
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ajay Sheshadri
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
2731
|
Antonelli A, Ferrari SM, Fallahi P. Current and future immunotherapies for thyroid cancer. Expert Rev Anticancer Ther 2017; 18:149-159. [PMID: 29241377 DOI: 10.1080/14737140.2018.1417845] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Cancer immunotherapies were approved in recent years, including immune checkpoint inhibitors. Experience with ipilimumab (CTLA-4 antagonist), nivolumab and pembrolizumab (PD-1 antagonists), and atezolizumab (PD-L1 antagonist) has shown that the impact on overall survival in cancer patients is paramount. Immune checkpoint inhibitors target the immune system and they can be applied across multiple cancers; the response rate is ranging from 20 to 40%. Many studies have shown that thyroid cancer (TC) cells produce cytokines and chemokines, inducing several tumor-promoting effects. Targeting and/or lowering cytokines and chemokines concentrations within the tumor microenvironment would produce a therapeutic benefit. In TC, increased Treg and PD-1+ T cell frequencies are indicative of aggressive disease and PD-L1 expression correlates with a greater risk of recurrence. Area covered: After performing a literature search, a few pioneering studies have evaluated immunotherapy in thyroid cancer. More recently a case has been described involving anaplastic thyroid cancer treated with vemurafenib and nivolumab, with substantial regression and complete radiographic and clinical remission. Expert commentary: The use of immune checkpoint inhibitors in aggressive TC has not yet been extensively investigated and further studies in a large number of TC patients are urgently needed.
Collapse
Affiliation(s)
- Alessandro Antonelli
- a Department of Clinical and Experimental Medicine , University of Pisa , Pisa , Italy
| | | | - Poupak Fallahi
- a Department of Clinical and Experimental Medicine , University of Pisa , Pisa , Italy
| |
Collapse
|
2732
|
Affiliation(s)
| | | | - James Larkin
- Royal Marsden NHS Foundation Trust, London, United Kingdom
| |
Collapse
|
2733
|
Abstract
This 'What's new in oncodermatology?' addresses the developments in 2017 on the epidemiology and management of skin cancers. We observe a constant increase in carcinomas, risk factors for squamous cell carcinoma, especially in transplant recipients where skin cancer mortality is important. Among epidemiological developments in melanoma are increased mortality despite screening, occupational exposure to UV, second melanoma and higher risk after carcinoma. New classifications that should be considered are AJCC8 for melanoma and carcinoma. In a near future artificial intelligence could change skin cancer screening practices through deep learning. For the sentinel lymph node, there is no interest in systematic lymphadenectomy that does not improve survival. Radiation therapy is essential for the prognosis of Merkel's carcinoma, and Mohs' surgery can be of interest. In metastatic melanoma, results on immunotherapy and targeted treatments include duration, dose, combinations, and the study of resistance mechanisms. The great novelty is immunotherapy or targeted therapy as an adjuvant treatment, giving an improvement in survival without relapse.
Collapse
Affiliation(s)
- C Lebbé
- AP-HP dermatologie, INSERM U976, université Paris 7-Diderot, hôpital Saint-Louis, Paris, France.
| |
Collapse
|
2734
|
Ikeda M, Morizane C, Ueno M, Okusaka T, Ishii H, Furuse J. Chemotherapy for hepatocellular carcinoma: current status and future perspectives. Jpn J Clin Oncol 2017; 48:103-114. [DOI: 10.1093/jjco/hyx180] [Citation(s) in RCA: 133] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 11/24/2017] [Indexed: 12/12/2022] Open
Affiliation(s)
- Masafumi Ikeda
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital East, Kashiwa
| | - Chigusa Morizane
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Tokyo
| | - Makoto Ueno
- Division of Hepatobiliary and Pancreatic Medical Oncology, Kanagawa Cancer Center, Yokohama
| | - Takuji Okusaka
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Tokyo
| | - Hiroshi Ishii
- Clinical Research Center, Shikoku Cancer Center, Matsuyama
| | - Junji Furuse
- Department of Medical Oncology, Kyorin University School of Medicine, Tokyo, Japan
| |
Collapse
|
2735
|
Stroh M. Designing In and Around Tolerability Considerations for Immunotherapy Combinations. Clin Pharmacol Ther 2017; 103:558-561. [DOI: 10.1002/cpt.945] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Mark Stroh
- CytomX Therapeutics Inc.South San Francisco California USA
| |
Collapse
|
2736
|
Abstract
Inhibiting the protein PD-1 can activate T cells that trigger immune responses against tumour cells. But it emerges that, in mice, this immunotherapy exacerbates a cancer that involves the T cells themselves.
Collapse
Affiliation(s)
- Aya Ludin
- the Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts 02138, USA
| | - Leonard I Zon
- the Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts 02138, USA
- the Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital and Dana-Farber Cancer Institute, Boston, Massachusetts, and at the Howard Hughes Medical Institute, Boston
| |
Collapse
|
2737
|
Torphy RJ, Schulick RD, Zhu Y. Newly Emerging Immune Checkpoints: Promises for Future Cancer Therapy. Int J Mol Sci 2017; 18:ijms18122642. [PMID: 29211042 PMCID: PMC5751245 DOI: 10.3390/ijms18122642] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 12/03/2017] [Accepted: 12/05/2017] [Indexed: 12/16/2022] Open
Abstract
Cancer immunotherapy has been a great breakthrough, with immune checkpoint inhibitors leading the way. Despite the clinical effectiveness of certain immune checkpoint inhibitors, the overall response rate remains low, and the effectiveness of immunotherapies for many tumors has been disappointing. There is substantial interest in looking for additional immune checkpoint molecules that may act as therapeutic targets for cancer. Recent advances during the last decade have identified several novel immune checkpoint targets, including lymphocyte activation gene-3 (LAG-3), B and T lymphocyte attenuator (BTLA), programmed death-1 homolog (PD-1H), T-cell immunoglobulin and immunoreceptor tyrosine-based inhibitory motif domain (TIM-3)/carcinoembryonic antigen cell adhesion molecule 1 (CEACAM1), and the poliovirus receptor (PVR)-like receptors. The investigations into these molecules have generated promising results in preclinical studies. Herein, we will summarize our current progress and understanding of these newly-characterized immune checkpoints and their potential application in cancer immunotherapy.
Collapse
Affiliation(s)
- Robert J Torphy
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.
| | - Richard D Schulick
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.
| | - Yuwen Zhu
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.
| |
Collapse
|
2738
|
Xu-Monette ZY, Zhang M, Li J, Young KH. PD-1/PD-L1 Blockade: Have We Found the Key to Unleash the Antitumor Immune Response? Front Immunol 2017; 8:1597. [PMID: 29255458 PMCID: PMC5723106 DOI: 10.3389/fimmu.2017.01597] [Citation(s) in RCA: 217] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 11/06/2017] [Indexed: 12/13/2022] Open
Abstract
PD-1-PD-L1 interaction is known to drive T cell dysfunction, which can be blocked by anti-PD-1/PD-L1 antibodies. However, studies have also shown that the function of the PD-1-PD-L1 axis is affected by the complex immunologic regulation network, and some CD8+ T cells can enter an irreversible dysfunctional state that cannot be rescued by PD-1/PD-L1 blockade. In most advanced cancers, except Hodgkin lymphoma (which has high PD-L1/L2 expression) and melanoma (which has high tumor mutational burden), the objective response rate with anti-PD-1/PD-L1 monotherapy is only ~20%, and immune-related toxicities and hyperprogression can occur in a small subset of patients during PD-1/PD-L1 blockade therapy. The lack of efficacy in up to 80% of patients was not necessarily associated with negative PD-1 and PD-L1 expression, suggesting that the roles of PD-1/PD-L1 in immune suppression and the mechanisms of action of antibodies remain to be better defined. In addition, important immune regulatory mechanisms within or outside of the PD-1/PD-L1 network need to be discovered and targeted to increase the response rate and to reduce the toxicities of immune checkpoint blockade therapies. This paper reviews the major functional and clinical studies of PD-1/PD-L1, including those with discrepancies in the pathologic and biomarker role of PD-1 and PD-L1 and the effectiveness of PD-1/PD-L1 blockade. The goal is to improve understanding of the efficacy of PD-1/PD-L1 blockade immunotherapy, as well as enhance the development of therapeutic strategies to overcome the resistance mechanisms and unleash the antitumor immune response to combat cancer.
Collapse
Affiliation(s)
- Zijun Y. Xu-Monette
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Mingzhi Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jianyong Li
- Department of Hematology, JiangSu Province Hospital, The First Affiliated Hospital of NanJing Medical University, NanJing, JiangSu Province, China
| | - Ken H. Young
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- Graduate School of Biomedical Science, The University of Texas Health Science Center at Houston, Houston, TX, United States
| |
Collapse
|
2739
|
Morello S, Capone M, Sorrentino C, Giannarelli D, Madonna G, Mallardo D, Grimaldi AM, Pinto A, Ascierto PA. Soluble CD73 as biomarker in patients with metastatic melanoma patients treated with nivolumab. J Transl Med 2017; 15:244. [PMID: 29202855 PMCID: PMC5716054 DOI: 10.1186/s12967-017-1348-8] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2017] [Accepted: 11/21/2017] [Indexed: 12/21/2022] Open
Abstract
Background Nivolumab is an anti-PD1 checkpoint inhibitor active in patients with advanced melanoma and as adjuvant therapy in high-risk metastatic melanoma patients. Methods In this single-center retrospective analysis, we investigated the CD73 enzyme activity in patients with metastatic melanoma stage IV and its correlation with the response to nivolumab. The soluble CD73 (sCD73) enzyme activity was measured in the serum of 37 melanoma patients before receiving nivolumab and the Harrel’s C index was used to find the best cut-off for this biomarker. The multivariate Cox proportional hazard model was used to evaluate the prognostic value of CD73 enzyme activity for survival and progression-free survival. Results Our results show that high levels of sCD73 enzyme activity were significantly associated with poor overall survival and progression-free survival in patients with metastatic melanoma. The median progression–free survival was 2.6 months [95% confidence interval (CI) 1.9–3.3] in patients with high sCD73 enzyme activity (> 27.8 pmol/min/mg protein), and 14.2 months (95% CI 4.6–23.8) in patients with lower CD73 enzyme activity, when patients were follow-up for a median of 24 months range. The median overall survival was not reached in patients with low sCD73 activity (< 27.8 pmol/min/mg protein) compared with 6.1 months (95% CI 0–14.8) in patients with higher sCD73 activity. In multivariate analyses, the sCD73 enzyme activity emerged as the strongest prognostic factor for overall survival and progression-free survival. Elevated basal levels of sCD73 enzyme activity, before starting nivolumab treatment, were associated with lower response rates to therapy. Conclusions We observed a significant association between the activity of sCD73 in the blood and clinical outcomes in patients with metastatic melanoma stage IV, receiving nivolumab. Although our results need to be confirmed and validated, we suggest that sCD73 might be used as serologic prognostic biomarker. Potentially evaluating sCD73 enzyme activity in the peripheral blood before treatment could help to estimate the response to nivolumab.
Collapse
Affiliation(s)
- Silvana Morello
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084, Fisciano, SA, Italy.
| | - Mariaelena Capone
- Melanoma, Cancer Immunotherapy and Innovative Therapies O.U, National Cancer Institute "G. Pascale", Naples, Italy
| | - Claudia Sorrentino
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084, Fisciano, SA, Italy.,PhD Program in Drug Discovery and Development, University of Salerno, Fisciano, SA, Italy
| | | | - Gabriele Madonna
- Melanoma, Cancer Immunotherapy and Innovative Therapies O.U, National Cancer Institute "G. Pascale", Naples, Italy
| | - Domenico Mallardo
- Melanoma, Cancer Immunotherapy and Innovative Therapies O.U, National Cancer Institute "G. Pascale", Naples, Italy
| | - Antonio M Grimaldi
- Melanoma, Cancer Immunotherapy and Innovative Therapies O.U, National Cancer Institute "G. Pascale", Naples, Italy
| | - Aldo Pinto
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084, Fisciano, SA, Italy
| | - Paolo Antonio Ascierto
- Melanoma, Cancer Immunotherapy and Innovative Therapies O.U, National Cancer Institute "G. Pascale", Naples, Italy
| |
Collapse
|
2740
|
Affiliation(s)
- Margarita Majem
- Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Charles M Rudin
- Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
2741
|
Liu B, Song Y, Liu D. Recent development in clinical applications of PD-1 and PD-L1 antibodies for cancer immunotherapy. J Hematol Oncol 2017; 10:174. [PMID: 29195503 PMCID: PMC5712158 DOI: 10.1186/s13045-017-0541-9] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Accepted: 11/15/2017] [Indexed: 12/16/2022] Open
Abstract
Antibodies against programmed death (PD) pathway are revolutionizing cancer immunotherapy. Currently five antibodies against PD-1/PD-L1 have been approved. The clinical use of these antibodies is rapidly expanding. Incorporation of PD antibodies into chemotherapy regimens is in active clinical investigations. The combination of pembrolizumab with carboplatin and pemetrexed has been approved for the first line therapy of metastatic non-squamous non-small cell lung cancer. Combination of PD-1/PD-L1 antibodies with small molecule inhibitors such as tyrosine kinase inhibitors and IDO inhibitors are in active clinical trials. This review summarized recent development in clinical trials of PD-1 and PD-L1 antibodies for cancer immunotherapy.
Collapse
Affiliation(s)
- Bingshan Liu
- School of Basic Medical Sciences and the Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
- Henan Cancer Hospital and the Affiliated Cancer Hospital of Zhengzhou University, 127 Dongming Road, Zhengzhou, 450008 China
| | - Yongping Song
- Henan Cancer Hospital and the Affiliated Cancer Hospital of Zhengzhou University, 127 Dongming Road, Zhengzhou, 450008 China
| | - Delong Liu
- Henan Cancer Hospital and the Affiliated Cancer Hospital of Zhengzhou University, 127 Dongming Road, Zhengzhou, 450008 China
| |
Collapse
|
2742
|
Dillman RO. An update on the relevance of vaccine research for the treatment of metastatic melanoma. Melanoma Manag 2017; 4:203-215. [PMID: 30190926 PMCID: PMC6094615 DOI: 10.2217/mmt-2017-0021] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 11/03/2017] [Indexed: 01/17/2023] Open
Abstract
Signal transduction inhibitors and anticheckpoint antibodies have significantly improved survival for metastatic melanoma patients, but most still die within 5 years. Vaccine approaches to induce immunity to well-characterized melanoma-associated antigens, or to antigens expressed on allogeneic tumor cell lines, have not resulted in approved agents. Despite the limitations associated with the immunosuppressive tumor microenvironment, there now is one intralesional autologous vaccine approved for patients who have primarily soft-tissue metastases. There is continued interest in patient-specific vaccines, especially dendritic cell vaccines that utilize ex vivo loading of autologous antigen, thus bypassing certain in vivo immunosuppressive cells and cytokines. Because of their mechanism of action and limited toxicity, they are potentially synergistic or additive to other antimelanoma therapies.
Collapse
Affiliation(s)
- Robert O Dillman
- Chief Medical Officer, AIVITA Biomedical, Inc; Clinical Professor of Medicine, University of California Irvine, Irvine, CA 92612, USA
| |
Collapse
|
2743
|
Davies MA, Flaherty KT. Melanoma in 2017: Moving treatments earlier to move further forwards. Nat Rev Clin Oncol 2017; 15:75-76. [PMID: 29182162 DOI: 10.1038/nrclinonc.2017.183] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Michael A Davies
- Department of Melanoma Medical Oncology, University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 0430, Houston, Texas 77030, USA
| | - Keith T Flaherty
- Center for Targeted Therapies, Massachusetts General Hospital Cancer Center, 55 Fruit Street, Boston, Massachusetts 02114, USA
| |
Collapse
|
2744
|
Jamal R, Lapointe R, Cocolakis E, Thébault P, Kazemi S, Friedmann JE, Dionne J, Cailhier JF, Bélanger K, Ayoub JP, Le H, Lambert C, El-Hajjar J, van Kempen LC, Spatz A, Miller WH. Peripheral and local predictive immune signatures identified in a phase II trial of ipilimumab with carboplatin/paclitaxel in unresectable stage III or stage IV melanoma. J Immunother Cancer 2017; 5:83. [PMID: 29157311 PMCID: PMC5696743 DOI: 10.1186/s40425-017-0290-x] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 10/04/2017] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Checkpoint blockade with ipilimumab provides long-term survival to a significant proportion of patients with metastatic melanoma. New approaches to increase survival and to predict which patients will benefit from treatment are needed. This phase II trial combined ipilimumab with carboplatin/paclitaxel (CP) to assess its safety, efficacy, and to search for peripheral and tumor-based predictive biomarkers. METHODS Thirty patients with untreated unresectable/metastatic melanoma were treated with ipilimumab and CP. Adverse events (AEs) were monitored and response to treatment was evaluated. Tumor tissue and peripheral blood were collected at specified time points to characterize tumor immune markers by immunohistochemistry and systemic immune activity by multiplex assays and flow cytometry. RESULTS Eighty three percent of patients received all 5 cycles of CP and 93% completed ipilimumab induction. Serious AEs occurred in 13% of patients, and no treatment-related deaths were observed. Best Overall Response Rate (BORR) and Disease Control Rate (DCR) were 27 and 57%, respectively. Median overall survival was 16.2 months. Response to treatment was positively correlated with a higher tumor CD3+ infiltrate (immune score) at baseline. NRAS and BRAF mutations were less frequent in patients who experienced clinical benefit. Assessment of peripheral blood revealed that non-responders had elevated baseline levels of CXCL8 and CCL4, and a higher proportion of circulating late differentiated B cells. Pre-existing high levels of chemokines (CCL3, CCL4 and CXCL8) and advanced B cell differentiation were strongly associated with worse patient overall survival. Elevated proportions of circulating CD8+/PD-1+ T cells during treatment were associated with worse survival. CONCLUSIONS The combination of ipilimumab and CP was well tolerated and revealed novel characteristics associated with patients likely to benefit from treatment. A pre-existing systemic inflammatory state characterized by elevation of selected chemokines and advanced B cell differentiation, was strongly associated with poor patient outcomes, revealing potential predictive circulating biomarkers. TRIAL REGISTRATION Clinicaltrials.gov , NCT01676649 , registered on August 29, 2012.
Collapse
Affiliation(s)
- Rahima Jamal
- Hôpital Notre-Dame, Centre de recherche du CHUM, Centre hospitalier de l’Université de Montréal, Montréal, QC Canada
| | - Réjean Lapointe
- Centre de recherche du CHUM, Institut du Cancer de Montréal, Université de Montréal, Montréal, QC Canada
| | - Eftihia Cocolakis
- Segal Cancer Center, Jewish General Hospital, Rossy Cancer Network, McGill University, 3755 Côte-St-Catherine, suite E670, Montreal, Québec Canada
| | - Paméla Thébault
- Centre de recherche du CHUM, Institut du Cancer de Montréal, Université de Montréal, Montréal, QC Canada
| | - Shirin Kazemi
- Segal Cancer Center, Jewish General Hospital, Rossy Cancer Network, McGill University, 3755 Côte-St-Catherine, suite E670, Montreal, Québec Canada
| | - Jennifer E. Friedmann
- Segal Cancer Center, Jewish General Hospital, Rossy Cancer Network, McGill University, 3755 Côte-St-Catherine, suite E670, Montreal, Québec Canada
| | - Jeanne Dionne
- Hôpital Notre-Dame, Centre de recherche du CHUM, Centre hospitalier de l’Université de Montréal, Montréal, QC Canada
| | - Jean-François Cailhier
- Centre de recherche du CHUM, Institut du Cancer de Montréal, Université de Montréal, Montréal, QC Canada
| | - Karl Bélanger
- Hôpital Notre-Dame, Centre de recherche du CHUM, Centre hospitalier de l’Université de Montréal, Montréal, QC Canada
| | - Jean-Pierre Ayoub
- Hôpital Notre-Dame, Centre de recherche du CHUM, Centre hospitalier de l’Université de Montréal, Montréal, QC Canada
| | - Huy Le
- Segal Cancer Center, Jewish General Hospital, Rossy Cancer Network, McGill University, 3755 Côte-St-Catherine, suite E670, Montreal, Québec Canada
| | - Caroline Lambert
- Segal Cancer Center, Jewish General Hospital, Rossy Cancer Network, McGill University, 3755 Côte-St-Catherine, suite E670, Montreal, Québec Canada
| | - Jida El-Hajjar
- Segal Cancer Center, Jewish General Hospital, Rossy Cancer Network, McGill University, 3755 Côte-St-Catherine, suite E670, Montreal, Québec Canada
| | - Léon C. van Kempen
- Department of Pathology, Molecular Pathology Center, Jewish General Hospital, McGill University, Montreal, QC Canada
| | - Alan Spatz
- Department of Pathology, Molecular Pathology Center, Jewish General Hospital, McGill University, Montreal, QC Canada
| | - Wilson H. Miller
- Segal Cancer Center, Jewish General Hospital, Rossy Cancer Network, McGill University, 3755 Côte-St-Catherine, suite E670, Montreal, Québec Canada
| |
Collapse
|
2745
|
Testa U, Castelli G, Pelosi E. Melanoma: Genetic Abnormalities, Tumor Progression, Clonal Evolution and Tumor Initiating Cells. Med Sci (Basel) 2017; 5:E28. [PMID: 29156643 PMCID: PMC5753657 DOI: 10.3390/medsci5040028] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2017] [Revised: 10/31/2017] [Accepted: 11/08/2017] [Indexed: 12/11/2022] Open
Abstract
Melanoma is an aggressive neoplasia issued from the malignant transformation of melanocytes, the pigment-generating cells of the skin. It is responsible for about 75% of deaths due to skin cancers. Melanoma is a phenotypically and molecularly heterogeneous disease: cutaneous, uveal, acral, and mucosal melanomas have different clinical courses, are associated with different mutational profiles, and possess distinct risk factors. The discovery of the molecular abnormalities underlying melanomas has led to the promising improvement of therapy, and further progress is expected in the near future. The study of melanoma precursor lesions has led to the suggestion that the pathway of tumor evolution implies the progression from benign naevi, to dysplastic naevi, to melanoma in situ and then to invasive and metastatic melanoma. The gene alterations characterizing melanomas tend to accumulate in these precursor lesions in a sequential order. Studies carried out in recent years have, in part, elucidated the great tumorigenic potential of melanoma tumor cells. These findings have led to speculation that the cancer stem cell model cannot be applied to melanoma because, in this malignancy, tumor cells possess an intrinsic plasticity, conferring the capacity to initiate and maintain the neoplastic process to phenotypically different tumor cells.
Collapse
Affiliation(s)
- Ugo Testa
- Department of Oncology, Istituto Superiore di Sanità, 00161 Rome, Italy.
| | - Germana Castelli
- Department of Oncology, Istituto Superiore di Sanità, 00161 Rome, Italy.
| | - Elvira Pelosi
- Department of Oncology, Istituto Superiore di Sanità, 00161 Rome, Italy.
| |
Collapse
|
2746
|
Marra A, Scognamiglio G, Peluso I, Botti G, Fusciello C, Filippelli A, Ascierto PA, Pepe S, Sabbatino F. Immune Checkpoint Inhibitors in Melanoma and HIV Infection. Open AIDS J 2017; 11:91-100. [PMID: 29290886 PMCID: PMC5730951 DOI: 10.2174/1874613601711010091] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Revised: 10/10/2017] [Accepted: 10/13/2017] [Indexed: 12/16/2022] Open
Abstract
Introduction: Immunotherapy with immune checkpoint inhibitors increases the overall survival of patients with metastatic melanoma regardless of their oncogene addicted mutations. However, no data is available from clinical trials of effective therapies in subgroups of melanoma patients that carry chronic infective diseases such as HIV. Evidences suggest a key role of the immune checkpoint molecules as a mechanism of immune escape not only from melanoma but also from HIV host immune response. Conclusion: In this article, firstly, we will describe the role of the immune checkpoint molecules in HIV chronic infection. Secondly, we will summarize the most relevant clinical evidences utilizing immune checkpoint inhibitors for the treatment of melanoma patients. Lastly, we will discuss the potential implications as well as the potential applications of immune checkpoint molecule-based immunotherapy in patients with melanoma and HIV infection.
Collapse
Affiliation(s)
- Antonio Marra
- Department of Medical Oncology, San Gerardo Hospital, via G. B. Pergolesi, 20052 Monza, Italy
| | - Giosuè Scognamiglio
- Pathology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione G. Pascale", via M. Semmola, 80131 Naples, Italy
| | - Ilaria Peluso
- Hematology Unit, Department of Clinical and Surgical Medicine, University of Naples Federico II, via S. Pansini, 80131 Naples, Italy
| | - Gerardo Botti
- Pathology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione G. Pascale", via M. Semmola, 80131 Naples, Italy
| | - Celeste Fusciello
- Oncology Unit, Department of Medicine, Surgery and Dentistry, University of Salerno, via Allende, 84081 Baronissi (Salerno), Italy
| | - Amelia Filippelli
- Pharmacology Unit, Department of Medicine, Surgery and Dentistry, University of Salerno, via Allende, 84081 Baronissi (Salerno), Italy
| | - Paolo A Ascierto
- Unit of Melanoma, Cancer Immunotherapy and Innovative Therapy, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione G. Pascale", via M. Semmola, 80131 Naples, Italy
| | - Stefano Pepe
- Oncology Unit, Department of Medicine, Surgery and Dentistry, University of Salerno, via Allende, 84081 Baronissi (Salerno), Italy
| | - Francesco Sabbatino
- Oncology Unit, Department of Medicine, Surgery and Dentistry, University of Salerno, via Allende, 84081 Baronissi (Salerno), Italy
| |
Collapse
|
2747
|
Robert C. Checkpoint Blockade Plus Oncolytic Virus: A Hot Therapeutic Cancer Strategy. Trends Mol Med 2017; 23:983-985. [DOI: 10.1016/j.molmed.2017.09.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 09/23/2017] [Indexed: 01/25/2023]
|
2748
|
Kostine M, Briaire-de Bruijn IH, Cleven AHG, Vervat C, Corver WE, Schilham MW, Van Beelen E, van Boven H, Haas RL, Italiano A, Cleton-Jansen AM, Bovée JVMG. Increased infiltration of M2-macrophages, T-cells and PD-L1 expression in high grade leiomyosarcomas supports immunotherapeutic strategies. Oncoimmunology 2017; 7:e1386828. [PMID: 29308311 PMCID: PMC5749622 DOI: 10.1080/2162402x.2017.1386828] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 09/25/2017] [Accepted: 09/26/2017] [Indexed: 10/25/2022] Open
Abstract
Background: Immunotherapy may be a rational strategy in leiomyosarcoma (LMS), a tumor known for its genomic complexity. As a prerequisite for therapeutic applications, we characterized the immune microenvironment in LMS, as well as its prognostic value. Methods: CD163+ macrophages, CD3+ T-cells, PD-L1/PD-L2 and HLA class I expression (HCA2, HC10 and β2m) were evaluated using immunohistochemistry in primary tumors (n = 75), local relapses (n = 6) and metastases (n = 19) of 87 LMS patients, as well as in benign leiomyomas (n = 7). Correlation with clinicopathological parameters and survival analyses were assessed. Effect of LMS cells on macrophage differentiation was investigated using coculture of CD14+ monocytes with LMS cell lines or their conditioned media (CM). Results: 58% and 52% of the tumors were highly infiltrated with CD163+ macrophages and T-cells, respectively, with HLA class I expression observed in almost all tumors and PD-L1 expression in 30%. PD-L2 expression was also detected in some PD-L1+ tumors. All these immune markers correlated with high tumor grade but only CD163 associated with overall survival (p = 0.003) and disease-specific survival (p = 0.041). In vitro, CD163 was upregulated in the presence of LMS cells producing M-CSF, suggesting that this tumor drives macrophages towards the M2 phenotype. Conclusion: The clinical significance of M2 macrophages, possibly induced by LMS cell-secreted factors, suggests that 2/3 of high-grade LMS patients might benefit from macrophage-targeting agents. Furthermore, PD-L1 expression together with high T-cell infiltrate and HLA class I expression in around 30% of high grade LMS reflects an active immune microenvironment potentially responsive to immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Marie Kostine
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands.,Department of Rheumatology, Hôpital Pellegrin, Place Amélie Raba Léon, Bordeaux, France
| | | | - Arjen H G Cleven
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Carly Vervat
- Department of Pediatrics, Leiden University Medical Center, Leiden, The Netherlands
| | - Willem E Corver
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Marco W Schilham
- Department of Pediatrics, Leiden University Medical Center, Leiden, The Netherlands
| | - Els Van Beelen
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Albinusdreef 2, ZA Leiden, The Netherlands
| | - Hester van Boven
- Department of Pathology, Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, The Netherlands
| | - Rick L Haas
- Department of Radiotherapy, Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, The Netherlands.,Department of Radiotherapy, Leiden University Medical Center, Leiden, The Netherlands
| | - Antoine Italiano
- Department of Medical Oncology, Institut Bergonié, Bordeaux, France
| | | | - Judith V M G Bovée
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
2749
|
Callahan MK, Kluger H, Postow MA, Segal NH, Lesokhin A, Atkins MB, Kirkwood JM, Krishnan S, Bhore R, Horak C, Wolchok JD, Sznol M. Nivolumab Plus Ipilimumab in Patients With Advanced Melanoma: Updated Survival, Response, and Safety Data in a Phase I Dose-Escalation Study. J Clin Oncol 2017; 36:391-398. [PMID: 29040030 DOI: 10.1200/jco.2017.72.2850] [Citation(s) in RCA: 139] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Purpose The clinical activity observed in a phase I dose-escalation study of concurrent therapy with nivolumab (NIVO) and ipilimumab (IPI) in patients with previously treated or untreated advanced melanoma led to subsequent clinical development, including randomized trials. Here, we report long-term follow-up data from study CA209-004, including 3-year overall survival (OS). Patients and Methods Concurrent cohorts 1, 2, 2a, and 3 received escalating doses of NIVO plus IPI once every 3 weeks for four doses, followed by NIVO once every 3 weeks for four doses, then NIVO plus IPI once every 12 weeks for eight doses. An expansion cohort (cohort 8) received concurrent NIVO 1 mg/kg plus IPI 3 mg/kg once every 3 weeks for four doses, followed by NIVO 3 mg/kg once every 2 weeks, which is the dose and schedule used in phase II and III studies and now approved for patients with unresectable or metastatic melanoma. Results Among all concurrent cohorts (N = 94) at a follow-up of 30.3 to 55.0 months, the 3-year OS rate was 63% and median OS had not been reached. Objective response rate by modified WHO criteria was 42%, and median duration of response was 22.3 months. Incidence of grade 3 and 4 treatment-related adverse events was 59%. The most common grade 3 and 4 treatment-related adverse events were increases in lipase (15%), alanine aminotransferase (12%), and aspartate aminotransferase (11%). One treatment-related death (1.1%) occurred in a patient who had multiorgan failure 70 days after the last dose of NIVO plus IPI. Conclusion This is the longest follow-up for NIVO plus IPI combination therapy in patients with advanced melanoma. The 3-year OS rate of 63% is the highest observed for this patient population and provides additional evidence for the durable clinical activity of immune checkpoint inhibitors in the treatment of advanced melanoma.
Collapse
Affiliation(s)
- Margaret K Callahan
- Margaret K. Callahan, Michael A. Postow, Neil H. Segal, Alexander Lesokhin, and Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center, and Weill Cornell Medical College, New York, NY; Harriet Kluger and Mario Sznol, Yale University School of Medicine and Smilow Cancer Center, and Yale-New Haven Hospital, New Haven, CT; Michael B. Atkins, Georgetown-Lombardi Comprehensive Cancer Center, Washington, DC; John M. Kirkwood, University of Pittsburgh Medical Center, Pittsburgh, PA; and Suba Krishnan, Rafia Bhore, and Christine Horak, Bristol-Myers Squibb, Princeton, NJ
| | - Harriet Kluger
- Margaret K. Callahan, Michael A. Postow, Neil H. Segal, Alexander Lesokhin, and Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center, and Weill Cornell Medical College, New York, NY; Harriet Kluger and Mario Sznol, Yale University School of Medicine and Smilow Cancer Center, and Yale-New Haven Hospital, New Haven, CT; Michael B. Atkins, Georgetown-Lombardi Comprehensive Cancer Center, Washington, DC; John M. Kirkwood, University of Pittsburgh Medical Center, Pittsburgh, PA; and Suba Krishnan, Rafia Bhore, and Christine Horak, Bristol-Myers Squibb, Princeton, NJ
| | - Michael A Postow
- Margaret K. Callahan, Michael A. Postow, Neil H. Segal, Alexander Lesokhin, and Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center, and Weill Cornell Medical College, New York, NY; Harriet Kluger and Mario Sznol, Yale University School of Medicine and Smilow Cancer Center, and Yale-New Haven Hospital, New Haven, CT; Michael B. Atkins, Georgetown-Lombardi Comprehensive Cancer Center, Washington, DC; John M. Kirkwood, University of Pittsburgh Medical Center, Pittsburgh, PA; and Suba Krishnan, Rafia Bhore, and Christine Horak, Bristol-Myers Squibb, Princeton, NJ
| | - Neil H Segal
- Margaret K. Callahan, Michael A. Postow, Neil H. Segal, Alexander Lesokhin, and Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center, and Weill Cornell Medical College, New York, NY; Harriet Kluger and Mario Sznol, Yale University School of Medicine and Smilow Cancer Center, and Yale-New Haven Hospital, New Haven, CT; Michael B. Atkins, Georgetown-Lombardi Comprehensive Cancer Center, Washington, DC; John M. Kirkwood, University of Pittsburgh Medical Center, Pittsburgh, PA; and Suba Krishnan, Rafia Bhore, and Christine Horak, Bristol-Myers Squibb, Princeton, NJ
| | - Alexander Lesokhin
- Margaret K. Callahan, Michael A. Postow, Neil H. Segal, Alexander Lesokhin, and Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center, and Weill Cornell Medical College, New York, NY; Harriet Kluger and Mario Sznol, Yale University School of Medicine and Smilow Cancer Center, and Yale-New Haven Hospital, New Haven, CT; Michael B. Atkins, Georgetown-Lombardi Comprehensive Cancer Center, Washington, DC; John M. Kirkwood, University of Pittsburgh Medical Center, Pittsburgh, PA; and Suba Krishnan, Rafia Bhore, and Christine Horak, Bristol-Myers Squibb, Princeton, NJ
| | - Michael B Atkins
- Margaret K. Callahan, Michael A. Postow, Neil H. Segal, Alexander Lesokhin, and Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center, and Weill Cornell Medical College, New York, NY; Harriet Kluger and Mario Sznol, Yale University School of Medicine and Smilow Cancer Center, and Yale-New Haven Hospital, New Haven, CT; Michael B. Atkins, Georgetown-Lombardi Comprehensive Cancer Center, Washington, DC; John M. Kirkwood, University of Pittsburgh Medical Center, Pittsburgh, PA; and Suba Krishnan, Rafia Bhore, and Christine Horak, Bristol-Myers Squibb, Princeton, NJ
| | - John M Kirkwood
- Margaret K. Callahan, Michael A. Postow, Neil H. Segal, Alexander Lesokhin, and Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center, and Weill Cornell Medical College, New York, NY; Harriet Kluger and Mario Sznol, Yale University School of Medicine and Smilow Cancer Center, and Yale-New Haven Hospital, New Haven, CT; Michael B. Atkins, Georgetown-Lombardi Comprehensive Cancer Center, Washington, DC; John M. Kirkwood, University of Pittsburgh Medical Center, Pittsburgh, PA; and Suba Krishnan, Rafia Bhore, and Christine Horak, Bristol-Myers Squibb, Princeton, NJ
| | - Suba Krishnan
- Margaret K. Callahan, Michael A. Postow, Neil H. Segal, Alexander Lesokhin, and Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center, and Weill Cornell Medical College, New York, NY; Harriet Kluger and Mario Sznol, Yale University School of Medicine and Smilow Cancer Center, and Yale-New Haven Hospital, New Haven, CT; Michael B. Atkins, Georgetown-Lombardi Comprehensive Cancer Center, Washington, DC; John M. Kirkwood, University of Pittsburgh Medical Center, Pittsburgh, PA; and Suba Krishnan, Rafia Bhore, and Christine Horak, Bristol-Myers Squibb, Princeton, NJ
| | - Rafia Bhore
- Margaret K. Callahan, Michael A. Postow, Neil H. Segal, Alexander Lesokhin, and Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center, and Weill Cornell Medical College, New York, NY; Harriet Kluger and Mario Sznol, Yale University School of Medicine and Smilow Cancer Center, and Yale-New Haven Hospital, New Haven, CT; Michael B. Atkins, Georgetown-Lombardi Comprehensive Cancer Center, Washington, DC; John M. Kirkwood, University of Pittsburgh Medical Center, Pittsburgh, PA; and Suba Krishnan, Rafia Bhore, and Christine Horak, Bristol-Myers Squibb, Princeton, NJ
| | - Christine Horak
- Margaret K. Callahan, Michael A. Postow, Neil H. Segal, Alexander Lesokhin, and Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center, and Weill Cornell Medical College, New York, NY; Harriet Kluger and Mario Sznol, Yale University School of Medicine and Smilow Cancer Center, and Yale-New Haven Hospital, New Haven, CT; Michael B. Atkins, Georgetown-Lombardi Comprehensive Cancer Center, Washington, DC; John M. Kirkwood, University of Pittsburgh Medical Center, Pittsburgh, PA; and Suba Krishnan, Rafia Bhore, and Christine Horak, Bristol-Myers Squibb, Princeton, NJ
| | - Jedd D Wolchok
- Margaret K. Callahan, Michael A. Postow, Neil H. Segal, Alexander Lesokhin, and Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center, and Weill Cornell Medical College, New York, NY; Harriet Kluger and Mario Sznol, Yale University School of Medicine and Smilow Cancer Center, and Yale-New Haven Hospital, New Haven, CT; Michael B. Atkins, Georgetown-Lombardi Comprehensive Cancer Center, Washington, DC; John M. Kirkwood, University of Pittsburgh Medical Center, Pittsburgh, PA; and Suba Krishnan, Rafia Bhore, and Christine Horak, Bristol-Myers Squibb, Princeton, NJ
| | - Mario Sznol
- Margaret K. Callahan, Michael A. Postow, Neil H. Segal, Alexander Lesokhin, and Jedd D. Wolchok, Memorial Sloan Kettering Cancer Center, and Weill Cornell Medical College, New York, NY; Harriet Kluger and Mario Sznol, Yale University School of Medicine and Smilow Cancer Center, and Yale-New Haven Hospital, New Haven, CT; Michael B. Atkins, Georgetown-Lombardi Comprehensive Cancer Center, Washington, DC; John M. Kirkwood, University of Pittsburgh Medical Center, Pittsburgh, PA; and Suba Krishnan, Rafia Bhore, and Christine Horak, Bristol-Myers Squibb, Princeton, NJ
| |
Collapse
|
2750
|
Control of NK Cell Activation by Immune Checkpoint Molecules. Int J Mol Sci 2017; 18:ijms18102129. [PMID: 29023417 PMCID: PMC5666811 DOI: 10.3390/ijms18102129] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 10/04/2017] [Accepted: 10/09/2017] [Indexed: 12/12/2022] Open
Abstract
The development of cancer and chronic infections is facilitated by many subversion mechanisms, among which enhanced expression of immune checkpoints molecules, such as programmed death-1 (PD-1) and cytotoxic T lymphocyte-associated antigen 4 (CTLA-4), on exhausted T cells. Recently, immune checkpoint inhibitors have shown remarkable efficiency in the treatment of a number of cancers. However, expression of immune checkpoints on natural killer (NK) cells and its functional consequences on NK cell effector functions are much less explored. In this review, we focus on the current knowledge on expression of various immune checkpoints in NK cells, how it can alter NK cell-mediated cytotoxicity and cytokine production. Dissecting the role of these inhibitory mechanisms in NK cells is critical for the full understanding of the mode of action of immunotherapies using checkpoint inhibitors in the treatment of cancers and chronic infections.
Collapse
|