5751
|
Shreders A, Joseph R, Peng C, Ye F, Zhao S, Puzanov I, Sosman JA, Johnson DB. Prolonged Benefit from Ipilimumab Correlates with Improved Outcomes from Subsequent Pembrolizumab. Cancer Immunol Res 2016; 4:569-73. [PMID: 27197063 PMCID: PMC4940026 DOI: 10.1158/2326-6066.cir-15-0281] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 03/12/2016] [Indexed: 11/16/2022]
Abstract
Patients with metastatic melanoma whose disease progresses on ipilimumab can clearly derive benefit from subsequent anti-programmed death-1 (PD-1). However, patients experience heterogeneous outcomes with ipilimumab, including rapid or delayed progression, and it is unclear whether patterns of ipilimumab progression influence subsequent clinical responses to anti-PD-1. We retrospectively reviewed data from 116 patients with metastatic melanoma who progressed on ipilimumab and were subsequently treated with pembrolizumab. The study objectives were to determine whether progression-free survival (PFS) with ipilimumab was associated with PFS, objective response rate (ORR), and clinical benefit rate (CBR; ORR + stable disease) with pembrolizumab. Patients with PFS ≥90 days after treatment with ipilimumab generally had superior outcomes with subsequent pembrolizumab treatment compared with patients with PFS <90 days (ORR, 49% vs. 35%, P = 0.12; CBR, 66% vs. 46%, P = 0.03). Patients with prolonged ipilimumab benefit (PFS ≥ 180 days) had excellent outcomes with pembrolizumab compared with rapid progressors (PFS < 45 days; ORR, 55% vs. 25%; CBR, 80% vs. 25%; median PFS, 249 vs. 50 days). Using logistic regression models, PFS with ipilimumab was independently correlated with response to pembrolizumab (odds ratio, 1.22; 95% CI, 1.02-1.51). This study shows that prolonged PFS with ipilimumab predicts excellent outcomes with subsequent pembrolizumab treatment, offering valuable prognostic information for clinicians. Cancer Immunol Res; 4(7); 569-73. ©2016 AACR.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal, Humanized/adverse effects
- Antibodies, Monoclonal, Humanized/drug effects
- Antineoplastic Agents, Immunological/administration & dosage
- Antineoplastic Agents, Immunological/adverse effects
- Antineoplastic Agents, Immunological/therapeutic use
- CTLA-4 Antigen/antagonists & inhibitors
- Combined Modality Therapy
- Drug Administration Schedule
- Female
- Humans
- Ipilimumab/administration & dosage
- Ipilimumab/adverse effects
- Ipilimumab/therapeutic use
- Male
- Melanoma/drug therapy
- Melanoma/mortality
- Melanoma/pathology
- Middle Aged
- Molecular Targeted Therapy
- Neoplasm Metastasis
- Programmed Cell Death 1 Receptor/antagonists & inhibitors
- Survival Analysis
- Treatment Outcome
- Young Adult
Collapse
Affiliation(s)
- Amanda Shreders
- Division of Hematology/Oncology, Department of Medicine, Mayo Clinic, Jacksonville, Florida.
| | - Richard Joseph
- Division of Hematology/Oncology, Department of Medicine, Mayo Clinic, Jacksonville, Florida
| | - Chengwei Peng
- Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Fei Ye
- Department of Quantitative Sciences, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Shilin Zhao
- Department of Quantitative Sciences, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Igor Puzanov
- Division of Hematology/Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Jeffrey A Sosman
- Division of Hematology/Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Douglas B Johnson
- Division of Hematology/Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
5752
|
Tartari F, Santoni M, Burattini L, Mazzanti P, Onofri A, Berardi R. Economic sustainability of anti-PD-1 agents nivolumab and pembrolizumab in cancer patients: Recent insights and future challenges. Cancer Treat Rev 2016; 48:20-24. [PMID: 27310708 DOI: 10.1016/j.ctrv.2016.06.002] [Citation(s) in RCA: 112] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 05/30/2016] [Accepted: 06/01/2016] [Indexed: 12/19/2022]
Abstract
Anti-programmed death (PD)-1 agents pembrolizumab and nivolumab have recently obtained enthusiastic results in terms of progression-free survival (PFS), overall survival (OS) and tolerability in cancer patients. Despite these promising data, the high cost of these agents needs careful consideration. Indeed, the evaluation of cost-effectiveness analysis (CEA) and quality-adjusted life year (QALY), as well as different drug reimbursement modalities, will represent fundamental tools in order to guarantee the economic sustainability of health system and the access to care for all cancer patients. In this review, we discussed the recent results obtained by immunotherapy in cancer patients and we evaluated the economic impact of recently approved nivolumab and pembrolizumab in patients with advanced melanoma, non-small cell lung cancer (NSCLC) and renal cell carcinoma (RCC).
Collapse
Affiliation(s)
| | - Matteo Santoni
- Medical Oncology, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria Ospedali Riuniti Umberto I, GM Lancisi, G Salesi, Ancona, Italy.
| | - Luciano Burattini
- Medical Oncology, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria Ospedali Riuniti Umberto I, GM Lancisi, G Salesi, Ancona, Italy
| | - Paola Mazzanti
- Medical Oncology, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria Ospedali Riuniti Umberto I, GM Lancisi, G Salesi, Ancona, Italy
| | - Azzurra Onofri
- Medical Oncology, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria Ospedali Riuniti Umberto I, GM Lancisi, G Salesi, Ancona, Italy
| | - Rossana Berardi
- Medical Oncology, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria Ospedali Riuniti Umberto I, GM Lancisi, G Salesi, Ancona, Italy
| |
Collapse
|
5753
|
Davies M. How Checkpoint Inhibitors Are Changing the Treatment Paradigm in Solid Tumors: What Advanced Practitioners in Oncology Need to Know. J Adv Pract Oncol 2016; 7:498-509. [PMID: 29282426 PMCID: PMC5737398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The immune system plays an active role in controlling and eradicating cancer. T cells, an essential component of the adaptive immune system, have a number of surface receptors (called "checkpoints") that can help either to sustain activation or suppress T-cell function. Many malignancies have developed ways to exploit these receptors to suppress T-cell function, enabling them to continue to grow. Anticancer immunotherapy in general, and checkpoint inhibitor therapy specifically, is a unique approach to cancer treatment that strives to harness the body's own immune system to generate an adequate response against cancer cells. Several checkpoint inhibitors are approved for the treatment of metastatic melanoma, non-small cell lung cancer, and renal cell carcinoma. These and other agents in this class are being investigated for their safety and efficacy in a variety of solid and hematologic malignancies. Advanced practitioners (APs) play a critical role in caring for patients treated with checkpoint inhibitors. It is essential for APs to be aware of the mechanism of action of these agents, patterns of response seen with this type of therapy, and presentation of immune-related adverse events related to these agents to ensure timely and successful treatment. Rapid evaluation/diagnostics and treatment are essential for optimal management and prevention of end-organ disease, and treatment of immune-related adverse effects requires a multidisciplinary approach.
Collapse
Affiliation(s)
- Marianne Davies
- Yale University School of Nursing, Yale Cancer Center, New Haven, Connecticut
| |
Collapse
|
5754
|
|
5755
|
Abstract
Over the last decade, the treatment of metastatic melanoma has been revolutionized by the translation of molecular insights into therapeutic benefit for patients. These include advances in immunotherapeutic and small-molecule approaches aimed at destroying cells with immunogenic antigens or gene mutations. Despite these advances, the limited durability of clinical response and eventual disease progression underscores a need for better understanding of mechanisms underlying tumor development. Current targeted therapies are developed partly based on the rationale that tumors are primarily clonal with respect to mutant oncogene or cell surface antigen target. However, with the advancement of cell isolation and transplantation approaches coupled with deep sequencing and mutation detection techniques, it has become increasingly clear that tumors are polyclonal. As a result, sensitive malignant cells are eradicated by treatment while the remaining tumor cell populations are conferred varying degrees of resistance and survival advantages by harbouring or acquiring certain epigenetic and genetic abnormalities. Tumor heterogeneity thus represents a major obstacle to the successful application of current therapies. Gaining insights into the cellular and molecular aspects of tumor diversity will not only facilitate the development and selection of therapeutic targets but also promote the evolution of precision medicine. In this viewpoint, we will discuss the implications of tumor heterogeneity for the treatment of metastatic melanoma and propose approaches to accelerate the translation of scientific discovery into improved clinical outcomes.
Collapse
Affiliation(s)
- Stephanie J. Hachey
- Department of Molecular Biology & Biochemistry, Sue & Bill Gross Stem Cell Research Center, CIRM Institute, University of California –Irvine, Irvine, CA 92697
| | - Alexander D. Boiko
- Department of Molecular Biology & Biochemistry, Sue & Bill Gross Stem Cell Research Center, CIRM Institute, University of California –Irvine, Irvine, CA 92697
- Correspondence should be addressed to Alexander D. Boiko ()
| |
Collapse
|
5756
|
Wicki A, Mandalà M, Massi D, Taverna D, Tang H, Hemmings BA, Xue G. Acquired Resistance to Clinical Cancer Therapy: A Twist in Physiological Signaling. Physiol Rev 2016; 96:805-29. [DOI: 10.1152/physrev.00024.2015] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Although modern therapeutic strategies have brought significant progress to cancer care in the last 30 years, drug resistance to targeted monotherapies has emerged as a major challenge. Aberrant regulation of multiple physiological signaling pathways indispensable for developmental and metabolic homeostasis, such as hyperactivation of pro-survival signaling axes, loss of suppressive regulations, and impaired functionalities of the immune system, have been extensively investigated aiming to understand the diversity of molecular mechanisms that underlie cancer development and progression. In this review, we intend to discuss the molecular mechanisms of how conventional physiological signal transduction confers to acquired drug resistance in cancer patients. We will particularly focus on protooncogenic receptor kinase inhibition-elicited tumor cell adaptation through two major core downstream signaling cascades, the PI3K/Akt and MAPK pathways. These pathways are crucial for cell growth and differentiation and are frequently hyperactivated during tumorigenesis. In addition, we also emphasize the emerging roles of the deregulated host immune system that may actively promote cancer progression and attenuate immunosurveillance in cancer therapies. Understanding these mechanisms may help to develop more effective therapeutic strategies that are able to keep the tumor in check and even possibly turn cancer into a chronic disease.
Collapse
Affiliation(s)
- Andreas Wicki
- Department of Biomedicine, University Hospital Basel, Basel, Switzerland; Department of Oncology and Hematology, Papa Giovanni XXIII Hospital, Bergamo, Italy; Department of Surgery and Translational Medicine, University of Florence, Florence, Italy; Department of Molecular Biotechnology and Health Sciences, University of Turin, Torino, Italy; Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou, China; and Department of Mechanisms of Cancer, Friedrich Miescher Institute for
| | - Mario Mandalà
- Department of Biomedicine, University Hospital Basel, Basel, Switzerland; Department of Oncology and Hematology, Papa Giovanni XXIII Hospital, Bergamo, Italy; Department of Surgery and Translational Medicine, University of Florence, Florence, Italy; Department of Molecular Biotechnology and Health Sciences, University of Turin, Torino, Italy; Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou, China; and Department of Mechanisms of Cancer, Friedrich Miescher Institute for
| | - Daniela Massi
- Department of Biomedicine, University Hospital Basel, Basel, Switzerland; Department of Oncology and Hematology, Papa Giovanni XXIII Hospital, Bergamo, Italy; Department of Surgery and Translational Medicine, University of Florence, Florence, Italy; Department of Molecular Biotechnology and Health Sciences, University of Turin, Torino, Italy; Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou, China; and Department of Mechanisms of Cancer, Friedrich Miescher Institute for
| | - Daniela Taverna
- Department of Biomedicine, University Hospital Basel, Basel, Switzerland; Department of Oncology and Hematology, Papa Giovanni XXIII Hospital, Bergamo, Italy; Department of Surgery and Translational Medicine, University of Florence, Florence, Italy; Department of Molecular Biotechnology and Health Sciences, University of Turin, Torino, Italy; Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou, China; and Department of Mechanisms of Cancer, Friedrich Miescher Institute for
| | - Huifang Tang
- Department of Biomedicine, University Hospital Basel, Basel, Switzerland; Department of Oncology and Hematology, Papa Giovanni XXIII Hospital, Bergamo, Italy; Department of Surgery and Translational Medicine, University of Florence, Florence, Italy; Department of Molecular Biotechnology and Health Sciences, University of Turin, Torino, Italy; Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou, China; and Department of Mechanisms of Cancer, Friedrich Miescher Institute for
| | - Brian A. Hemmings
- Department of Biomedicine, University Hospital Basel, Basel, Switzerland; Department of Oncology and Hematology, Papa Giovanni XXIII Hospital, Bergamo, Italy; Department of Surgery and Translational Medicine, University of Florence, Florence, Italy; Department of Molecular Biotechnology and Health Sciences, University of Turin, Torino, Italy; Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou, China; and Department of Mechanisms of Cancer, Friedrich Miescher Institute for
| | - Gongda Xue
- Department of Biomedicine, University Hospital Basel, Basel, Switzerland; Department of Oncology and Hematology, Papa Giovanni XXIII Hospital, Bergamo, Italy; Department of Surgery and Translational Medicine, University of Florence, Florence, Italy; Department of Molecular Biotechnology and Health Sciences, University of Turin, Torino, Italy; Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou, China; and Department of Mechanisms of Cancer, Friedrich Miescher Institute for
| |
Collapse
|
5757
|
Miller KD, Siegel RL, Lin CC, Mariotto AB, Kramer JL, Rowland JH, Stein KD, Alteri R, Jemal A. Cancer treatment and survivorship statistics, 2016. CA Cancer J Clin 2016; 66:271-89. [PMID: 27253694 DOI: 10.3322/caac.21349] [Citation(s) in RCA: 3527] [Impact Index Per Article: 391.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The number of cancer survivors continues to increase because of both advances in early detection and treatment and the aging and growth of the population. For the public health community to better serve these survivors, the American Cancer Society and the National Cancer Institute collaborate to estimate the number of current and future cancer survivors using data from the Surveillance, Epidemiology, and End Results cancer registries. In addition, current treatment patterns for the most prevalent cancer types are presented based on information in the National Cancer Data Base and treatment-related side effects are briefly described. More than 15.5 million Americans with a history of cancer were alive on January 1, 2016, and this number is projected to reach more than 20 million by January 1, 2026. The 3 most prevalent cancers are prostate (3,306,760), colon and rectum (724,690), and melanoma (614,460) among males and breast (3,560,570), uterine corpus (757,190), and colon and rectum (727,350) among females. More than one-half (56%) of survivors were diagnosed within the past 10 years, and almost one-half (47%) are aged 70 years or older. People with a history of cancer have unique medical and psychosocial needs that require proactive assessment and management by primary care providers. Although there are a growing number of tools that can assist patients, caregivers, and clinicians in navigating the various phases of cancer survivorship, further evidence-based resources are needed to optimize care. CA Cancer J Clin 2016;66:271-289. © 2016 American Cancer Society.
Collapse
Affiliation(s)
- Kimberly D Miller
- Epidemiologist, Surveillance and Health Services Research, American Cancer Society, Atlanta, GA
| | - Rebecca L Siegel
- Strategic Director, Surveillance Information, Surveillance and Health Services Research, American Cancer Society, Atlanta, GA
| | - Chun Chieh Lin
- Director, Health Services Research, Intramural Research Department, American Cancer Society, Atlanta, GA
| | - Angela B Mariotto
- Branch Chief, Surveillance Research Program, National Cancer Institute, Bethesda, MD
| | - Joan L Kramer
- Assistant Professor, Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA
| | - Julia H Rowland
- Director, Office of Cancer Survivorship, National Cancer Institute, Bethesda, MD
| | - Kevin D Stein
- Vice President, Behavioral Research Center, American Cancer Society, Atlanta, GA
| | - Rick Alteri
- Medical Editor, American Cancer Society, Atlanta, GA
| | - Ahmedin Jemal
- Vice President, Surveillance and Health Services Research, American Cancer Society, Atlanta, GA
| |
Collapse
|
5758
|
Resistance to combination BRAF and MEK inhibition in metastatic melanoma: Where to next? Eur J Cancer 2016; 62:76-85. [DOI: 10.1016/j.ejca.2016.04.005] [Citation(s) in RCA: 148] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 04/05/2016] [Indexed: 12/12/2022]
|
5759
|
Strauss J, Madan RA, Gulley JL. Considerations for the combination of anticancer vaccines and immune checkpoint inhibitors. Expert Opin Biol Ther 2016; 16:895-901. [PMID: 27010190 PMCID: PMC6599515 DOI: 10.1517/14712598.2016.1170805] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
INTRODUCTION Over the past few years, trials evaluating immunotherapies, particularly immune checkpoint inhibitors, have revolutionized the standard model of cancer treatment, demonstrating significant antitumor responses and improved clinical outcomes across a wide array of tumors types. Yet, despite these compelling data, a major limitation has been that only a fraction of patients mount a response to single-agent immune checkpoint inhibition. However, a growing amount of preclinical and clinical data suggests that combining immune checkpoint inhibition, either with other immune checkpoint inhibitors or with therapeutic cancer vaccines, has the potential to improve the proportion of patients seeing long-term durable responses with these therapies. AREAS COVERED We have reviewed the reported data on immune checkpoint inhibition as monotherapy and as combination therapy with other immune checkpoint inhibitors or therapeutic cancer vaccines. Data is reviewed on agents with FDA approval or breakthrough designation as of the writing of this manuscript. EXPERT OPINION Particular focus is given to the combination of immune checkpoint inhibitors and therapeutic cancer vaccines which has the potential to increase efficacy compared to single agent immune checkpoint inhibition with minimal added toxicity.
Collapse
Affiliation(s)
- Julius Strauss
- a Genitourinary Malignancies Branch , Center for Cancer Research, National Cancer Institute , Bethesda , MD , USA
| | - Ravi A Madan
- a Genitourinary Malignancies Branch , Center for Cancer Research, National Cancer Institute , Bethesda , MD , USA
| | - James L Gulley
- a Genitourinary Malignancies Branch , Center for Cancer Research, National Cancer Institute , Bethesda , MD , USA
| |
Collapse
|
5760
|
Naylor EC. Adjuvant Therapy for Stage I and II Non–Small Cell Lung Cancer. Surg Oncol Clin N Am 2016; 25:585-99. [DOI: 10.1016/j.soc.2016.03.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
5761
|
Delconte RB, Kolesnik TB, Dagley LF, Rautela J, Shi W, Putz EM, Stannard K, Zhang JG, Teh C, Firth M, Ushiki T, Andoniou CE, Degli-Esposti MA, Sharp PP, Sanvitale CE, Infusini G, Liau NPD, Linossi EM, Burns CJ, Carotta S, Gray DHD, Seillet C, Hutchinson DS, Belz GT, Webb AI, Alexander WS, Li SS, Bullock AN, Babon JJ, Smyth MJ, Nicholson SE, Huntington ND. CIS is a potent checkpoint in NK cell-mediated tumor immunity. Nat Immunol 2016; 17:816-24. [PMID: 27213690 DOI: 10.1038/ni.3470] [Citation(s) in RCA: 281] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2016] [Accepted: 04/27/2016] [Indexed: 12/14/2022]
Abstract
The detection of aberrant cells by natural killer (NK) cells is controlled by the integration of signals from activating and inhibitory ligands and from cytokines such as IL-15. We identified cytokine-inducible SH2-containing protein (CIS, encoded by Cish) as a critical negative regulator of IL-15 signaling in NK cells. Cish was rapidly induced in response to IL-15, and deletion of Cish rendered NK cells hypersensitive to IL-15, as evidenced by enhanced proliferation, survival, IFN-γ production and cytotoxicity toward tumors. This was associated with increased JAK-STAT signaling in NK cells in which Cish was deleted. Correspondingly, CIS interacted with the tyrosine kinase JAK1, inhibiting its enzymatic activity and targeting JAK for proteasomal degradation. Cish(-/-) mice were resistant to melanoma, prostate and breast cancer metastasis in vivo, and this was intrinsic to NK cell activity. Our data uncover a potent intracellular checkpoint in NK cell-mediated tumor immunity and suggest possibilities for new cancer immunotherapies directed at blocking CIS function.
Collapse
Affiliation(s)
- Rebecca B Delconte
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Victoria, Australia
| | - Tatiana B Kolesnik
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Laura F Dagley
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Victoria, Australia
| | - Jai Rautela
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Victoria, Australia
| | - Wei Shi
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Victoria, Australia
| | - Eva M Putz
- Immunology in Cancer and Infection Laboratory QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Kimberley Stannard
- Immunology in Cancer and Infection Laboratory QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Jian-Guo Zhang
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Victoria, Australia
| | - Charis Teh
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Victoria, Australia
| | - Matt Firth
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Victoria, Australia
| | - Takashi Ushiki
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Victoria, Australia
| | - Christopher E Andoniou
- Immunology and Virology Program, Centre for Ophthalmology and Visual Science, The University of Western Australia and Centre for Experimental Immunology, Lions Eye Institute, Nedlands, Western Australia, Australia
| | - Mariapia A Degli-Esposti
- Immunology and Virology Program, Centre for Ophthalmology and Visual Science, The University of Western Australia and Centre for Experimental Immunology, Lions Eye Institute, Nedlands, Western Australia, Australia
| | - Phillip P Sharp
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Victoria, Australia
| | | | - Giuseppe Infusini
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Nicholas P D Liau
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Victoria, Australia
| | - Edmond M Linossi
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Victoria, Australia
| | - Christopher J Burns
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Victoria, Australia
| | - Sebastian Carotta
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Victoria, Australia
| | - Daniel H D Gray
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Victoria, Australia
| | - Cyril Seillet
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Victoria, Australia
| | - Dana S Hutchinson
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Gabrielle T Belz
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Victoria, Australia
| | - Andrew I Webb
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Victoria, Australia
| | - Warren S Alexander
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Victoria, Australia
| | - Shawn S Li
- Department of Biochemistry and the Siebens-Drake Medical Research Institute, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Alex N Bullock
- Structural Genomics Consortium (SGC), University of Oxford, Oxford, UK
| | - Jeffery J Babon
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Victoria, Australia
| | - Mark J Smyth
- Immunology in Cancer and Infection Laboratory QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
- School of Medicine, University of Queensland, Herston, Queensland, Australia
| | - Sandra E Nicholson
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Victoria, Australia
| | - Nicholas D Huntington
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Victoria, Australia
| |
Collapse
|
5762
|
Antonia SJ, López-Martin JA, Bendell J, Ott PA, Taylor M, Eder JP, Jäger D, Pietanza MC, Le DT, de Braud F, Morse MA, Ascierto PA, Horn L, Amin A, Pillai RN, Evans J, Chau I, Bono P, Atmaca A, Sharma P, Harbison CT, Lin CS, Christensen O, Calvo E. Nivolumab alone and nivolumab plus ipilimumab in recurrent small-cell lung cancer (CheckMate 032): a multicentre, open-label, phase 1/2 trial. Lancet Oncol 2016; 17:883-895. [PMID: 27269741 DOI: 10.1016/s1470-2045(16)30098-5] [Citation(s) in RCA: 992] [Impact Index Per Article: 110.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 04/07/2016] [Accepted: 04/20/2016] [Indexed: 02/08/2023]
Abstract
BACKGROUND Treatments for small-cell lung cancer (SCLC) after failure of platinum-based chemotherapy are limited. We assessed safety and activity of nivolumab and nivolumab plus ipilimumab in patients with SCLC who progressed after one or more previous regimens. METHODS The SCLC cohort of this phase 1/2 multicentre, multi-arm, open-label trial was conducted at 23 sites (academic centres and hospitals) in six countries. Eligible patients were 18 years of age or older, had limited-stage or extensive-stage SCLC, and had disease progression after at least one previous platinum-containing regimen. Patients received nivolumab (3 mg/kg bodyweight intravenously) every 2 weeks (given until disease progression or unacceptable toxicity), or nivolumab plus ipilimumab (1 mg/kg plus 1 mg/kg, 1 mg/kg plus 3 mg/kg, or 3 mg/kg plus 1 mg/kg, intravenously) every 3 weeks for four cycles, followed by nivolumab 3 mg/kg every 2 weeks. Patients were either assigned to nivolumab monotherapy or assessed in a dose-escalating safety phase for the nivolumab/ipilimumab combination beginning at nivolumab 1 mg/kg plus ipilimumab 1 mg/kg. Depending on tolerability, patients were then assigned to nivolumab 1 mg/kg plus ipilimumab 3 mg/kg or nivolumab 3 mg/kg plus ipilimumab 1 mg/kg. The primary endpoint was objective response by investigator assessment. All analyses included patients who were enrolled at least 90 days before database lock. This trial is ongoing; here, we report an interim analysis of the SCLC cohort. This study is registered with ClinicalTrials.gov, number NCT01928394. FINDINGS Between Nov 18, 2013, and July 28, 2015, 216 patients were enrolled and treated (98 with nivolumab 3 mg/kg, three with nivolumab 1 mg/kg plus ipilimumab 1 mg/kg, 61 with nivolumab 1 mg/kg plus ipilimumab 3 mg/kg, and 54 with nivolumab 3 mg/kg plus ipilimumab 1 mg/kg). At database lock on Nov 6, 2015, median follow-up for patients continuing in the study (including those who had died or discontinued treatment) was 198·5 days (IQR 163·0-464·0) for nivolumab 3 mg/kg, 302 days (IQR not calculable) for nivolumab 1 mg/kg plus ipilimumab 1 mg/kg, 361·0 days (273·0-470·0) for nivolumab 1 mg/kg plus ipilimumab 3 mg/kg, and 260·5 days (248·0-288·0) for nivolumab 3 mg/kg plus ipilimumab 1 mg/kg. An objective response was achieved in ten (10%) of 98 patients receiving nivolumab 3 mg/kg, one (33%) of three patients receiving nivolumab 1 mg/kg plus ipilimumab 1 mg/kg, 14 (23%) of 61 receiving nivolumab 1 mg/kg plus ipilimumab 3 mg/kg, and ten (19%) of 54 receiving nivolumab 3 mg/kg plus ipilimumab 1 mg/kg. Grade 3 or 4 treatment-related adverse events occurred in 13 (13%) patients in the nivolumab 3 mg/kg cohort, 18 (30%) in the nivolumab 1 mg/kg plus ipilimumab 3 mg/kg cohort, and ten (19%) in the nivolumab 3 mg/kg plus ipilimumab 1 mg/kg cohort; the most commonly reported grade 3 or 4 treatment-related adverse events were increased lipase (none vs 5 [8%] vs none) and diarrhoea (none vs 3 [5%] vs 1 [2%]). No patients in the nivolumab 1 mg/kg plus ipilimumab 1 mg/kg cohort had a grade 3 or 4 treatment-related adverse event. Six (6%) patients in the nivolumab 3 mg/kg group, seven (11%) in the nivolumab 1 mg/kg plus ipilimumab 3 mg/kg group, and four (7%) in the nivolumab 3 mg/kg plus ipilimumab 1 mg/kg group discontinued treatment due to treatment-related adverse events. Two patients who received nivolumab 1 mg/kg plus ipilimumab 3 mg/kg died from treatment-related adverse events (myasthenia gravis and worsening of renal failure), and one patient who received nivolumab 3 mg/kg plus ipilimumab 1 mg/kg died from treatment-related pneumonitis. INTERPRETATION Nivolumab monotherapy and nivolumab plus ipilimumab showed antitumour activity with durable responses and manageable safety profiles in previously treated patients with SCLC. These data suggest a potential new treatment approach for a population of patients with limited treatment options and support the evaluation of nivolumab and nivolumab plus ipilimumab in phase 3 randomised controlled trials in SCLC. FUNDING Bristol-Myers Squibb.
Collapse
Affiliation(s)
- Scott J Antonia
- H Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | | | - Johanna Bendell
- Sarah Cannon Research Institute/Tennessee Oncology, PLLC, Nashville, TN, USA
| | | | | | | | - Dirk Jäger
- Nationales Centrum für Tumorerkrankungen (NCT), University Medical Center, Heidelberg, Germany
| | | | - Dung T Le
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD, USA
| | - Filippo de Braud
- Fondazione IRCCS Istituto Nazionale dei Tumori Milano, Milan, Italy
| | | | | | - Leora Horn
- Vanderbilt-Ingram Cancer Center, Nashville, TN, USA
| | - Asim Amin
- Levine Cancer Institute, Carolinas Medical Center, Charlotte, NC, USA
| | - Rathi N Pillai
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | | | - Ian Chau
- Royal Marsden Hospital, Sutton, UK
| | - Petri Bono
- Comprehensive Cancer Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Akin Atmaca
- Krankenhaus Nordwest UCT-University Cancer Center, Frankfurt, Germany
| | - Padmanee Sharma
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | | | | | - Emiliano Calvo
- START Madrid, Centro Integral Oncológico Clara Campal, Madrid, Spain.
| |
Collapse
|
5763
|
Manson G, Norwood J, Marabelle A, Kohrt H, Houot R. Biomarkers associated with checkpoint inhibitors. Ann Oncol 2016; 27:1199-206. [DOI: 10.1093/annonc/mdw181] [Citation(s) in RCA: 119] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 04/18/2016] [Indexed: 02/07/2023] Open
|
5764
|
Krecké N, Zimmer A, Friesenhahn-Ochs B, Müller CSL, Vogt T, Pföhler C. Sneaky side effects and ineffectiveness of an immunotherapy with ipilimumab in a case of metastatic melanoma. DERMATO-ENDOCRINOLOGY 2016; 8:e1199307. [PMID: 27574531 PMCID: PMC4977105 DOI: 10.1080/19381980.2016.1199307] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 06/03/2016] [Indexed: 11/11/2022]
Abstract
Ipilimumab is an anti-CTLA-4 antibody that is approved for the treatment of metastatic malignant melanoma. Side-effects are mostly immune-mediated and in many cases the lack of specific symptoms leads to delayed diagnosis and treatment of adverse events. We present the case of a female patient who experienced an uncommon combination of adverse reactions while undergoing therapy with ipilimumab and where the absence of specificity of the symptoms led to late diagnosis and treatment of side effects. Autoimmune disease was neither associated with tumor response nor with prolonged survival.
Collapse
Affiliation(s)
- Nathalie Krecké
- Department of Dermatology, Saarland University Medical School , Homburg/Saar, Germany
| | - Anna Zimmer
- Department for Diagnostic and Interventional Neuroradiology, Saarland University Medical School , Homburg/Saar, Germany
| | | | - Cornelia S L Müller
- Department of Dermatology, Saarland University Medical School , Homburg/Saar, Germany
| | - Thomas Vogt
- Department of Dermatology, Saarland University Medical School , Homburg/Saar, Germany
| | - Claudia Pföhler
- Department of Dermatology, Saarland University Medical School , Homburg/Saar, Germany
| |
Collapse
|
5765
|
Kim JE, Patel MA, Mangraviti A, Kim ES, Theodros D, Velarde E, Liu A, Sankey EW, Tam A, Xu H, Mathios D, Jackson CM, Harris-Bookman S, Garzon-Muvdi T, Sheu M, Martin AM, Tyler BM, Tran PT, Ye X, Olivi A, Taube JM, Burger PC, Drake CG, Brem H, Pardoll DM, Lim M. Combination Therapy with Anti-PD-1, Anti-TIM-3, and Focal Radiation Results in Regression of Murine Gliomas. Clin Cancer Res 2016; 23:124-136. [PMID: 27358487 DOI: 10.1158/1078-0432.ccr-15-1535] [Citation(s) in RCA: 352] [Impact Index Per Article: 39.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2015] [Revised: 05/01/2016] [Accepted: 05/27/2016] [Indexed: 02/06/2023]
Abstract
PURPOSE Checkpoint molecules like programmed death-1 (PD-1) and T-cell immunoglobulin mucin-3 (TIM-3) are negative immune regulators that may be upregulated in the setting of glioblastoma multiforme. Combined PD-1 blockade and stereotactic radiosurgery (SRS) have been shown to improve antitumor immunity and produce long-term survivors in a murine glioma model. However, tumor-infiltrating lymphocytes (TIL) can express multiple checkpoints, and expression of ≥2 checkpoints corresponds to a more exhausted T-cell phenotype. We investigate TIM-3 expression in a glioma model and the antitumor efficacy of TIM-3 blockade alone and in combination with anti-PD-1 and SRS. EXPERIMENTAL DESIGN C57BL/6 mice were implanted with murine glioma cell line GL261-luc2 and randomized into 8 treatment arms: (i) control, (ii) SRS, (iii) anti-PD-1 antibody, (iv) anti-TIM-3 antibody, (v) anti-PD-1 + SRS, (vi) anti-TIM-3 + SRS, (vii) anti-PD-1 + anti-TIM-3, and (viii) anti-PD-1 + anti-TIM-3 + SRS. Survival and immune activation were assessed. RESULTS Dual therapy with anti-TIM-3 antibody + SRS or anti-TIM-3 + anti-PD-1 improved survival compared with anti-TIM-3 antibody alone. Triple therapy resulted in 100% overall survival (P < 0.05), a significant improvement compared with other arms. Long-term survivors demonstrated increased immune cell infiltration and activity and immune memory. Finally, positive staining for TIM-3 was detected in 7 of 8 human GBM samples. CONCLUSIONS This is the first preclinical investigation on the effects of dual PD-1 and TIM-3 blockade with radiation. We also demonstrate the presence of TIM-3 in human glioblastoma multiforme and provide preclinical evidence for a novel treatment combination that can potentially result in long-term glioma survival and constitutes a novel immunotherapeutic strategy for the treatment of glioblastoma multiforme. Clin Cancer Res; 23(1); 124-36. ©2016 AACR.
Collapse
Affiliation(s)
- Jennifer E Kim
- Department of Neurosurgery, Johns Hopkins University, Baltimore, Maryland
| | - Mira A Patel
- Department of Neurosurgery, Johns Hopkins University, Baltimore, Maryland
| | | | - Eileen S Kim
- Department of Neurosurgery, Johns Hopkins University, Baltimore, Maryland
| | - Debebe Theodros
- Department of Neurosurgery, Johns Hopkins University, Baltimore, Maryland
| | - Esteban Velarde
- Department of Radiation Oncology, Johns Hopkins University, Baltimore, Maryland
| | - Ann Liu
- Department of Neurosurgery, Johns Hopkins University, Baltimore, Maryland
| | - Eric W Sankey
- Department of Neurosurgery, Johns Hopkins University, Baltimore, Maryland
| | - Ada Tam
- Flow Cytometry Core, Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland
| | - Haiying Xu
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland
| | - Dimitrios Mathios
- Department of Neurosurgery, Johns Hopkins University, Baltimore, Maryland
| | | | | | - Tomas Garzon-Muvdi
- Department of Neurosurgery, Johns Hopkins University, Baltimore, Maryland
| | - Mary Sheu
- Department of Dermatology, Johns Hopkins University, Baltimore, Maryland
| | - Allison M Martin
- Department of Neurosurgery, Johns Hopkins University, Baltimore, Maryland
| | - Betty M Tyler
- Department of Neurosurgery, Johns Hopkins University, Baltimore, Maryland
| | - Phuoc T Tran
- Department of Radiation Oncology, Johns Hopkins University, Baltimore, Maryland
| | - Xiaobu Ye
- Department of Neurosurgery, Johns Hopkins University, Baltimore, Maryland
| | - Alessandro Olivi
- Department of Neurosurgery, Johns Hopkins University, Baltimore, Maryland
| | - Janis M Taube
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland
| | - Peter C Burger
- Department of Neurosurgery, Johns Hopkins University, Baltimore, Maryland.,Department of Pathology, Johns Hopkins University, Baltimore, Maryland.,Department of Oncology, Johns Hopkins University, Baltimore, Maryland
| | - Charles G Drake
- Department of Oncology, Johns Hopkins University, Baltimore, Maryland
| | - Henry Brem
- Department of Neurosurgery, Johns Hopkins University, Baltimore, Maryland
| | - Drew M Pardoll
- Department of Oncology, Johns Hopkins University, Baltimore, Maryland
| | - Michael Lim
- Department of Neurosurgery, Johns Hopkins University, Baltimore, Maryland.
| |
Collapse
|
5766
|
Akiyama Y, Nonomura C, Kondou R, Miyata H, Ashizawa T, Maeda C, Mitsuya K, Hayashi N, Nakasu Y, Yamaguchi K. Immunological effects of the anti-programmed death-1 antibody on human peripheral blood mononuclear cells. Int J Oncol 2016; 49:1099-107. [PMID: 27573705 DOI: 10.3892/ijo.2016.3586] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 05/30/2016] [Indexed: 11/06/2022] Open
Abstract
Immune checkpoint antibody-mediated blockade has gained attention as a new cancer immunotherapy strategy. Accumulating evidence suggests that this therapy imparts a survival benefit to metastatic melanoma and non-small cell lung cancer patients. A substantial amount of data on immune checkpoint antibodies has been collected from clinical trials; however, the direct effect of the antibodies on human peripheral blood mononuclear cells (PBMCs) has not been exclusively investigated. In this study, we developed an anti-programmed death-1 (PD-1) antibody (with biosimilarity to nivolumab) and examined the effects of the antibody on PBMCs derived from cancer patients. Specifically, we investigated the effects of the anti-PD-1 antibody on proliferation, cytokine production, cytotoxic T lymphocytes (CTL) and regulatory T cells. These investigations yielded several important results. First, the anti-PD-1 antibody had no obvious effect on resting PBMCs; however, high levels of the anti-PD-1 antibody partly stimulated PBMC proliferation when accompanied by an anti-CD3 antibody. Second, the anti-PD-1 antibody restored the growth inhibition of anti-CD3 Ab-stimulated PBMCs mediated by PD-L1. Third, the anti-PD-1 antibody exhibited a moderate inhibitory effect on the induction of myeloid-derived suppressor cells (MDSCs) by anti-CD3 antibody stimulation. Additionally, the presence of the anti-PD-1 antibody promoted antigen-specific CTL induction, which suggests that combining anti-PD-1 antibody and conventional immunotherapy treatments may have beneficial effects. These results indicate that specific cellular immunological mechanisms are partly responsible for the antitumor effect exhibited by the anti-PD-1 antibody against advanced cancers in clinical trials.
Collapse
Affiliation(s)
- Yasuto Akiyama
- Immunotherapy Division, Shizuoka Cancer Center Research Institute, Nagaizumi-cho, Sunto-gun, Shizuoka 411-8777, Japan
| | - Chizu Nonomura
- Immunotherapy Division, Shizuoka Cancer Center Research Institute, Nagaizumi-cho, Sunto-gun, Shizuoka 411-8777, Japan
| | - Ryota Kondou
- Immunotherapy Division, Shizuoka Cancer Center Research Institute, Nagaizumi-cho, Sunto-gun, Shizuoka 411-8777, Japan
| | - Haruo Miyata
- Immunotherapy Division, Shizuoka Cancer Center Research Institute, Nagaizumi-cho, Sunto-gun, Shizuoka 411-8777, Japan
| | - Tadashi Ashizawa
- Immunotherapy Division, Shizuoka Cancer Center Research Institute, Nagaizumi-cho, Sunto-gun, Shizuoka 411-8777, Japan
| | - Chie Maeda
- Immunotherapy Division, Shizuoka Cancer Center Research Institute, Nagaizumi-cho, Sunto-gun, Shizuoka 411-8777, Japan
| | - Koichi Mitsuya
- Division of Neurosurgery, Shizuoka Cancer Center Hospital, Nagaizumi-cho, Sunto-gun, Shizuoka 411-8777, Japan
| | - Nakamasa Hayashi
- Division of Neurosurgery, Shizuoka Cancer Center Hospital, Nagaizumi-cho, Sunto-gun, Shizuoka 411-8777, Japan
| | - Yoko Nakasu
- Division of Neurosurgery, Shizuoka Cancer Center Hospital, Nagaizumi-cho, Sunto-gun, Shizuoka 411-8777, Japan
| | - Ken Yamaguchi
- Office of The President, Shizuoka Cancer Center Hospital, Nagaizumi-cho, Sunto-gun, Shizuoka 411-8777, Japan
| |
Collapse
|
5767
|
Chae YK, Arya A, Malecek MK, Shin DS, Carneiro B, Chandra S, Kaplan J, Kalyan A, Altman JK, Platanias L, Giles F. Repurposing metformin for cancer treatment: current clinical studies. Oncotarget 2016; 7:40767-40780. [PMID: 27004404 PMCID: PMC5130043 DOI: 10.18632/oncotarget.8194] [Citation(s) in RCA: 231] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2015] [Accepted: 03/06/2016] [Indexed: 12/13/2022] Open
Abstract
In recent years, several studies have presented evidence suggesting a potential role for metformin in anti-cancer therapy. Preclinical studies have demonstrated several anticancer molecular mechanisms of metformin including mTOR inhibition, cytotoxic effects, and immunomodulation. Epidemiologic data have demonstrated decreased cancer incidence and mortality in patients taking metformin. Several clinical trials, focused on evaluation of metformin as an anti-cancer agent are presently underway. Data published from a small number of completed trials has put forth intriguing results. Clinical trials in pre-surgical endometrial cancer patients exhibited a significant decrease in Ki67 with metformin monotherapy. Another interesting observation was made in patients with breast cancer, wherein a trend towards improvement in cancer proliferation markers was noted in patients without insulin resistance. Data on survival outcomes with the use of metformin as an anti-cancer agent is awaited. This manuscript will critically review the role of metformin as a potential cancer treatment.
Collapse
Affiliation(s)
- Young Kwang Chae
- Northwestern Medicine Developmental Therapeutics Institute, Chicago, IL, USA
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA
- Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Ayush Arya
- Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Mary-Kate Malecek
- Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Daniel Sanghoon Shin
- David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Benedito Carneiro
- Northwestern Medicine Developmental Therapeutics Institute, Chicago, IL, USA
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA
- Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Sunandana Chandra
- Northwestern Medicine Developmental Therapeutics Institute, Chicago, IL, USA
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA
- Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Jason Kaplan
- Northwestern Medicine Developmental Therapeutics Institute, Chicago, IL, USA
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA
- Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Aparna Kalyan
- Northwestern Medicine Developmental Therapeutics Institute, Chicago, IL, USA
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA
- Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Jessica K. Altman
- Northwestern Medicine Developmental Therapeutics Institute, Chicago, IL, USA
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA
- Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Leonidas Platanias
- Northwestern Medicine Developmental Therapeutics Institute, Chicago, IL, USA
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA
- Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Division of Hematology-Oncology, Department of Medicine, Jesse Brown VA Medical Center, Chicago, IL, USA
| | - Francis Giles
- Northwestern Medicine Developmental Therapeutics Institute, Chicago, IL, USA
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA
- Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
5768
|
Vreeland TJ, Clifton GT, Herbert GS, Hale DF, Jackson DO, Berry JS, Peoples GE. Gaining ground on a cure through synergy: combining checkpoint inhibitors with cancer vaccines. Expert Rev Clin Immunol 2016; 12:1347-1357. [PMID: 27323245 DOI: 10.1080/1744666x.2016.1202114] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION The approval of multiple checkpoint inhibitors (CPIs) for the treatment of advanced malignancies has sparked an explosion of research in the field of cancer immunotherapy. Despite the success of these medications, a large number of patients with advanced malignancy do not benefit from therapy. Early research indicates that a therapeutic combination of cancer vaccines with checkpoint inhibitors may lead to synergistic effects and higher response rates than monotherapy. Areas covered: This paper summarizes the previously completed and ongoing research on this exciting combination, including the use of the tumor lysate, particle-loaded dendritic cell (TLPLDC) vaccine combined with checkpoint inhibitors in advanced melanoma. Expert commentary: Increasing experience with CPIs has led to improved understanding of which patients may benefit and it is increasingly clear that the presence of a pre-existing immune response to the tumor, along with tumor-infiltrating lymphocytes, is key to the success of CPIs. One exciting possibility for the future is the addition of a cancer vaccine to CPI therapy, eliciting these crucial T cells, which can then be augmented and protected by the CPI. A number of current and future studies are addressing this very exciting combination therapy.
Collapse
Affiliation(s)
- T J Vreeland
- a Department of Surgery , Womack Army Medical Center , Fort Bragg , NC , USA
| | - G T Clifton
- b Department of Surgical Oncology , University of Texas MD Anderson Cancer Center , Houston , TX , USA
| | - G S Herbert
- c Department of Surgery , Brooke Army Medical Center , Fort Sam Houston , TX , USA
| | - D F Hale
- c Department of Surgery , Brooke Army Medical Center , Fort Sam Houston , TX , USA
| | - D O Jackson
- c Department of Surgery , Brooke Army Medical Center , Fort Sam Houston , TX , USA
| | - J S Berry
- c Department of Surgery , Brooke Army Medical Center , Fort Sam Houston , TX , USA
| | - G E Peoples
- b Department of Surgical Oncology , University of Texas MD Anderson Cancer Center , Houston , TX , USA.,d Cancer Vaccine Development Program, San Antonio, TX and Department of Surgery , Uniformed Services University of the Health Sciences , Bethesda , MD , USA
| |
Collapse
|
5769
|
Yeh I. Recent advances in molecular genetics of melanoma progression: implications for diagnosis and treatment. F1000Res 2016; 5. [PMID: 27408703 PMCID: PMC4926755 DOI: 10.12688/f1000research.8247.1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/22/2016] [Indexed: 11/20/2022] Open
Abstract
According to the multi-step carcinogenesis model of cancer, initiation results in a benign tumor and subsequent genetic alterations lead to tumor progression and the acquisition of the hallmarks of cancer. This article will review recent discoveries in our understanding of initiation and progression in melanocytic neoplasia and the impact on diagnostic dermatopathology.
Collapse
Affiliation(s)
- Iwei Yeh
- Department of Dermatology, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
5770
|
Hypoxic stress: obstacles and opportunities for innovative immunotherapy of cancer. Oncogene 2016; 36:439-445. [PMID: 27345407 DOI: 10.1038/onc.2016.225] [Citation(s) in RCA: 275] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 05/03/2016] [Accepted: 05/17/2016] [Indexed: 12/11/2022]
Abstract
Tumors use several strategies to evade the host immune response, including creation of an immune-suppressive and hostile tumor environment. Tissue hypoxia due to inadequate blood supply is reported to develop very early during tumor establishment. Hypoxic stress has a strong impact on tumor cell biology. In particular, tissue hypoxia contributes to therapeutic resistance, heterogeneity and progression. It also interferes with immune plasticity, promotes the differentiation and expansion of immune-suppressive stromal cells, and remodels the metabolic landscape to support immune privilege. Therefore, tissue hypoxia has been regarded as a central factor for tumor aggressiveness and metastasis. In this regard, manipulating host-tumor interactions in the context of the hypoxic tumor microenvironment may be important in preventing or reverting malignant conversion. We will discuss how tumor microenvironment-driven transient compositional tumor heterogeneity involves hypoxic stress. Tumor hypoxia is a therapeutic concern since it can reduce the effectiveness of conventional therapies as well as cancer immunotherapy. Thus, understanding how tumor and stromal cells respond to hypoxia will allow for the design of innovative cancer therapies that can overcome these barriers. A better understanding of hypoxia-dependent mechanisms involved in the regulation of immune tolerance could lead to new strategies to enhance antitumor immunity. Therefore, discovery and validation of therapeutic targets derived from the hypoxic tumor microenvironment is of major importance. In this context, critical hypoxia-associated pathways are attractive targets for immunotherapy of cancer. In this review, we summarize current knowledge regarding the molecular mechanisms induced by tumor cell hypoxia with a special emphasis on therapeutic resistance and immune suppression. We emphasize mechanisms of manipulating hypoxic stress and its associated pathways, which may support the development of more durable and successful cancer immunotherapy approaches in the future.
Collapse
|
5771
|
Ren Z, Guo J, Liao J, Luan Y, Liu Z, Sun Z, Liu X, Liang Y, Peng H, Fu YX. CTLA-4 Limits Anti-CD20-Mediated Tumor Regression. Clin Cancer Res 2016; 23:193-203. [PMID: 27354469 DOI: 10.1158/1078-0432.ccr-16-0040] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Revised: 06/12/2016] [Accepted: 06/15/2016] [Indexed: 01/25/2023]
Abstract
PURPOSE The inhibition of tumor growth by anti-CD20 antibody (Ab) treatment is mediated by Ab- and complement-dependent cytotoxicity in xenograft tumor models. In addition, anti-CD20 therapy for B-cell lymphoma can result in intrinsic and extrinsic tumor resistance to further Ab treatment. However, adaptive immune response-related resistance has not been well studied in anti-CD20-mediated tumor control, and adaptive immunity has long been underestimated. The purpose of this study was to explore whether T cells are involved in mediating the effects of anti-CD20 therapy and what factors contribute to adaptive immune response-related resistance. EXPERIMENTAL DESIGN Using a syngeneic mouse B-cell lymphoma model, we investigated the role of CD8+ T cells in anti-CD20-mediated tumor regression. Furthermore, we revealed how the tumor-specific T-cell response was initiated by anti-CD20. Finally, we studied adaptive immune response-related resistance in advanced B-cell lymphoma. RESULTS CD8+ T cells played an essential role in anti-CD20-mediated tumor regression. Mechanistically, anti-CD20 therapy promoted dendritic cell (DC)-mediated cross-presentation. Importantly, macrophages were also necessary for the increase in the tumor-specific CTL response after anti-CD20 treatment, via the production of type I IFN to activate DC function. Furthermore, adaptive resistance is gradually developed through the CTLA-4 pathway in Treg cells in larger lymphomas. Further blockade of CTLA-4 can synergize with anti-CD20 treatment in antitumor activities. CONCLUSIONS The therapeutic function of anti-CD20 depends on tumor-specific CD8+ T-cell responses initiated by anti-CD20 through macrophages and DCs. CTLA-4 blockade can synergize with anti-CD20 to overcome adaptive immune response-related resistance in advanced B-cell lymphoma. Clin Cancer Res; 23(1); 193-203. ©2016 AACR.
Collapse
Affiliation(s)
- Zhenhua Ren
- Key Laboratory of Infection and Immunity of CAS, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Jingya Guo
- Key Laboratory of Infection and Immunity of CAS, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Jing Liao
- Key Laboratory of Infection and Immunity of CAS, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Yan Luan
- Key Laboratory of Infection and Immunity of CAS, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,DingFu Biotarget Co. Ltd., Suzhou, Jiangsu, China
| | - Zhida Liu
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Zhichen Sun
- Key Laboratory of Infection and Immunity of CAS, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Xiaojuan Liu
- Key Laboratory of Infection and Immunity of CAS, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Yong Liang
- Key Laboratory of Infection and Immunity of CAS, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Hua Peng
- Key Laboratory of Infection and Immunity of CAS, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.
| | - Yang-Xin Fu
- Key Laboratory of Infection and Immunity of CAS, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China. .,Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
5772
|
Jensen CE, Loaiza-Bonilla A, Bonilla-Reyes PA. Immune checkpoint inhibitors for hepatocellular carcinoma. Hepat Oncol 2016; 3:201-211. [PMID: 30191042 DOI: 10.2217/hep-2016-0004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 04/25/2016] [Indexed: 12/19/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is a leading cause of cancer deaths worldwide, and advanced HCC generally caries a poor prognosis. The treatment of advanced disease is limited to sorafenib, which provides only a limited improvement in survival, and novel therapies are, thus, sorely needed. Among emerging alternative approaches, immune checkpoint inhibitors are a particularly promising treatment modality. In this review, we summarize current knowledge of the mechanisms for the two primary targets of immune checkpoint inhibitors and discuss the relevance of these pathways to the immunology of HCC. We also review the state of ongoing and forthcoming trials of immune checkpoint blockade in HCC.
Collapse
Affiliation(s)
- Christopher E Jensen
- Department of Medicine, The Hospital of the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA.,Department of Medicine, The Hospital of the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA
| | - Arturo Loaiza-Bonilla
- Departments of Medicine, Hematology & Oncology, Abramson Cancer Center of the University of Pennsylvania, Perelman Center for Advanced Medicine, 6th Floor South Pavilion, 3400 Civic Center Boulevard, Philadelphia, PA 19104, USA.,Departments of Medicine, Hematology & Oncology, Abramson Cancer Center of the University of Pennsylvania, Perelman Center for Advanced Medicine, 6th Floor South Pavilion, 3400 Civic Center Boulevard, Philadelphia, PA 19104, USA
| | - Paula A Bonilla-Reyes
- Facultad de Medicina, Pontificia Universidad Javeriana, Cra. 7 No. 40-62, Hospital Universitario, San Ignacio, Bogota, Colombia.,Facultad de Medicina, Pontificia Universidad Javeriana, Cra. 7 No. 40-62, Hospital Universitario, San Ignacio, Bogota, Colombia
| |
Collapse
|
5773
|
Rajkumar S, Watson IR. Molecular characterisation of cutaneous melanoma: creating a framework for targeted and immune therapies. Br J Cancer 2016; 115:145-55. [PMID: 27336610 PMCID: PMC4947706 DOI: 10.1038/bjc.2016.195] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Revised: 05/16/2016] [Accepted: 05/18/2016] [Indexed: 12/12/2022] Open
Abstract
Large-scale genomic analyses of cutaneous melanoma have revealed insights into the aetiology and heterogeneity of this disease, as well as opportunities to further personalise treatment for patients with targeted and immune therapies. Herein, we review the proposed genomic classification of cutaneous melanoma from large-scale next-generation sequencing studies, including the largest integrative analysis of melanoma from The Cancer Genome Atlas (TCGA) Network. We examine studies that have identified molecular features of melanomas linked to immune checkpoint inhibitor response. In addition, we draw attention to low-frequency actionable mutations and highlight frequent non-coding mutations in melanoma where little is known about their biological function that may provide novel avenues for the development of treatment strategies for melanoma patients.
Collapse
Affiliation(s)
- Shivshankari Rajkumar
- Rosalind and Morris Goodman Cancer Research Center, Department of Biochemistry, McGill University, Montréal, QC, Canada H3A 1A3
| | - Ian R Watson
- Rosalind and Morris Goodman Cancer Research Center, Department of Biochemistry, McGill University, Montréal, QC, Canada H3A 1A3
| |
Collapse
|
5774
|
Kaufman HL, Butterfield LH, Coulie PG, Demaria S, Ferris RL, Galon J, Khleif SN, Mellman I, Ohashi PS, Overwijk WW, Topalian SL, Marincola FM. Society for immunotherapy of cancer (SITC) statement on the proposed changes to the common rule. J Immunother Cancer 2016; 4:37. [PMID: 27330810 PMCID: PMC4915147 DOI: 10.1186/s40425-016-0142-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 06/08/2016] [Indexed: 12/04/2022] Open
Abstract
The Common Rule is a set of ethical principles that provide guidance on the management of human subjects taking part in biomedical and behavioral research in the United States. The elements of the Common Rule were initially developed in 1981 following a revision of the Declaration of Helsinki in 1975. Most academic facilities follow the Common Rule in the regulation of clinical trials research. Recently, the government has suggested a revision of the Common Rule to include more contemporary and streamlined oversight of clinical research. In this commentary, the leadership of the Society for Immunotherapy of Cancer (SITC) provides their opinion on this plan. While the Society recognizes the considerable contribution of clinical research in supporting progress in tumor immunotherapy and supports the need for revisions to the Common Rule, there is also some concern over certain elements which may restrict access to biospecimens and clinical data at a time when high throughput technologies, computational biology and assay standardization is allowing major advances in understanding cancer biology and providing potential predictive biomarkers of immunotherapy response. The Society values its professional commitment to patients for improving clinical outcomes with tumor immunotherapy and supports continued discussion with all stakeholders before implementing changes to the Common Rule in order to ensure maximal patient protections while promoting continued clinical research at this historic time in cancer research.
Collapse
Affiliation(s)
- Howard L Kaufman
- Rutgers Cancer Institute of New Jersey, 195 Little Albany Street, Room 2004, New Brunswick, NJ 08901 USA
| | | | - Pierre G Coulie
- De Duve Institute, Universite Catholique De Louvain, Brussels, Belgium
| | | | | | - Jérôme Galon
- Cordeliers Research Center, INSERMFrench National Institute of Health and Medical Research, Paris, France
| | - Samir N Khleif
- Georgia Regents University Cancer Center, Augusta, GA France
| | | | - Pamela S Ohashi
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | | | | | | |
Collapse
|
5775
|
Freeston S. Combining immune checkpoint inhibitors for advanced melanoma: the key to optimizing treatment outcomes? J Comp Eff Res 2016; 5:329-33. [PMID: 27331424 DOI: 10.2217/cer-2016-0023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
5776
|
Filippi AR, Fava P, Badellino S, Astrua C, Ricardi U, Quaglino P. Radiotherapy and immune checkpoints inhibitors for advanced melanoma. Radiother Oncol 2016; 120:1-12. [PMID: 27345592 DOI: 10.1016/j.radonc.2016.06.003] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 05/26/2016] [Accepted: 06/08/2016] [Indexed: 01/05/2023]
Abstract
INTRODUCTION The therapeutic landscape of metastatic melanoma drastically changed after the introduction of targeted therapies and immunotherapy, in particular immune checkpoints inhibitors (ICI). In recent years, positive effects on the immune system associated to radiotherapy (RT) were discovered, and radiation has been tested in combination with ICI in both pre-clinical and clinical studies (many of them still ongoing). We here summarize the rationale and the preliminary clinical results of this approach. MATERIALS AND METHODS In the first part of this review article, redacted with narrative non-systematic methodology, we describe the clinical results of immune checkpoints blockade in melanoma as well as the biological basis for the combination of ICI with RT; in the second part, we systematically review scientific publications reporting on the clinical results of the combination of ICI and RT for advanced melanoma. RESULTS The biological and mechanistic rationale behind the combination of ICI and radiation is well supported by several preclinical findings. Retrospective observational series and few prospective trials support the potential synergistic effect between radiation and ICI for metastatic melanoma. CONCLUSION RT may potentiate anti-melanoma activity of ICI by enhancing response on both target and non-target lesions. Several prospective trials are ongoing with the aim of further exploring this combination in the clinical setting, hopefully confirming initial observations and opening a new therapeutic window for advanced melanoma patients.
Collapse
Affiliation(s)
| | - Paolo Fava
- Department of Medical Sciences, Dermatology/Oncology, University of Torino, Italy
| | - Serena Badellino
- Department of Oncology, Radiation Oncology, University of Torino, Italy
| | - Chiara Astrua
- Department of Medical Sciences, Dermatology/Oncology, University of Torino, Italy
| | - Umberto Ricardi
- Department of Oncology, Radiation Oncology, University of Torino, Italy
| | - Pietro Quaglino
- Department of Medical Sciences, Dermatology/Oncology, University of Torino, Italy
| |
Collapse
|
5777
|
Delitto D, Wallet SM, Hughes SJ. Targeting tumor tolerance: A new hope for pancreatic cancer therapy? Pharmacol Ther 2016; 166:9-29. [PMID: 27343757 DOI: 10.1016/j.pharmthera.2016.06.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Accepted: 06/09/2016] [Indexed: 01/18/2023]
Abstract
With a 5-year survival rate of just 8%, pancreatic cancer (PC) is projected to be the second leading cause of cancer deaths by 2030. Most PC patients are not eligible for surgery with curative intent upon diagnosis, emphasizing a need for more effective therapies. However, PC is notoriously resistant to chemoradiation regimens. As an alternative, immune modulating strategies have recently achieved success in melanoma, prompting their application to other solid tumors. For such therapeutic approaches to succeed, a state of immunologic tolerance must be reversed in the tumor microenvironment and that has been especially challenging in PC. Nonetheless, knowledge of the PC immune microenvironment has advanced considerably over the past decade, yielding new insights and perspectives to guide multimodal therapies. In this review, we catalog the historical groundwork and discuss the evolution of the cancer immunology field to its present state with a specific focus on PC. Strategies currently employing immune modulation in PC are reviewed, specifically highlighting 66 clinical trials across the United States and Europe.
Collapse
Affiliation(s)
- Daniel Delitto
- Department of Surgery, University of Florida, Gainesville, FL, USA
| | - Shannon M Wallet
- Department of Oral Biology, University of Florida, Gainesville, FL, USA
| | - Steven J Hughes
- Department of Surgery, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
5778
|
Bilen MA, Subudhi SK, Gao J, Tannir NM, Tu SM, Sharma P. Acute rhabdomyolysis with severe polymyositis following ipilimumab-nivolumab treatment in a cancer patient with elevated anti-striated muscle antibody. J Immunother Cancer 2016; 4:36. [PMID: 27330809 PMCID: PMC4915058 DOI: 10.1186/s40425-016-0139-8] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 05/23/2016] [Indexed: 01/05/2023] Open
Abstract
Background Immune checkpoint inhibitors have revolutionized cancer therapy since these drugs target inhibitory pathways on T cells, which result in durable anti-tumor immune responses and significant overall survival for a subset of cancer patients. These drugs can also lead to toxicities, which require additional research to identify mechanisms of toxicities and biomarkers that can help to identify patients who will develop immune-related adverse events. Case presentation We describe the first case, to our knowledge, of a patient with metastatic urothelial carcinoma who developed acute rhabdomyolysis with severe polymyositis after treatment with combination immunotherapy consisting of ipilimumab plus nivolumab (Trial registration: NCT01928394. Registered: 8/21/2013). We found that this patient had an elevated pre-existing anti-striated muscle antibody titer, which was likely exacerbated with the immunotherapy treatment thereby resulting in the presentation of acute rhabdomyolysis and severe polymyositis. Conclusions This case suggests that immune-related adverse events may be linked to subclinical autoimmune conditions which highlights the need for additional studies to identify patients who are at risk for toxicities.
Collapse
Affiliation(s)
- Mehmet Asim Bilen
- Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Sumit K Subudhi
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Unit 1374, 1155 Pressler Street, Houston, TX 77030-3721 USA
| | - Jianjun Gao
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Unit 1374, 1155 Pressler Street, Houston, TX 77030-3721 USA
| | - Nizar M Tannir
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Unit 1374, 1155 Pressler Street, Houston, TX 77030-3721 USA
| | - Shi-Ming Tu
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Unit 1374, 1155 Pressler Street, Houston, TX 77030-3721 USA
| | - Padmanee Sharma
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Unit 1374, 1155 Pressler Street, Houston, TX 77030-3721 USA
| |
Collapse
|
5779
|
Shien K, Papadimitrakopoulou VA, Wistuba II. Predictive biomarkers of response to PD-1/PD-L1 immune checkpoint inhibitors in non-small cell lung cancer. Lung Cancer 2016; 99:79-87. [PMID: 27565919 DOI: 10.1016/j.lungcan.2016.06.016] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 06/13/2016] [Accepted: 06/19/2016] [Indexed: 02/07/2023]
Abstract
Inhibitors of the programmed cell death 1 (PD-1) pathway show the potential to substantially increase the efficacy of therapy for various malignancies, including non-small cell lung cancer (NSCLC). At the same time, substantial effort has been invested in finding biomarkers predicting which patients will respond best to this immune checkpoint inhibition. PD-L1 expression in tumor cells and the tumor microenvironment, genetic alterations and mutational load in tumor cells, and pre-existing immunity and its enhancement during treatment through tumor-infiltrating immune cells have been associated with outcomes of immune checkpoint inhibition. Here, we review the reported predictive biomarkers of response to PD-1 pathway immune checkpoint inhibitors in NSCLC, mainly focusing on results obtained with clinical trials.
Collapse
Affiliation(s)
- Kazuhiko Shien
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, 2130 W. Holcombe Blvd., Houston, TX 77030, USA
| | - Vassiliki A Papadimitrakopoulou
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA
| | - Ignacio I Wistuba
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, 2130 W. Holcombe Blvd., Houston, TX 77030, USA.
| |
Collapse
|
5780
|
Boudewijns S, Bloemendal M, Gerritsen WR, de Vries IJM, Schreibelt G. Dendritic cell vaccination in melanoma patients: From promising results to future perspectives. Hum Vaccin Immunother 2016; 12:2523-2528. [PMID: 27322496 DOI: 10.1080/21645515.2016.1197453] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Dendritic cells (DCs) play an important role in the induction of antitumor immunity. Therefore, they are used as anti-cancer vaccines in clinical studies in various types of cancer. DC vaccines are generally well tolerated and able to induce antigen-specific T cell responses in melanoma patients. After DC vaccinations, functional tumor-specific T cells are more frequently detected in stage III melanoma patients, as compared to patients with advanced melanoma, indicating that the tumor load influences immunological responses. Furthermore, long-lasting clinical responses were rarely seen in metastatic melanoma patients after DC vaccination. Since more potent treatment options are available, e.g. immune checkpoint inhibitors and targeted therapy, DC vaccination as monotherapy may not be preferred in the treatment of advanced melanoma. However, encouraging results of DC vaccines combined with ipilimumab have been reported in advanced melanoma patients with an objective response rate of 38%. DC vaccines show promising clinical results in stage III patients, although clinical efficacy still needs to be proven in a phase 3 trial. The clinical and immunological results of DC vaccination in stage III melanoma patients might be further improved by using naturally circulating DCs (myeloid DCs and plasmacytoid DCs) and neoantigens to load DCs.
Collapse
Affiliation(s)
- Steve Boudewijns
- a Department of Medical Oncology , Radboud University Medical Center , Nijmegen , The Netherlands.,b Department of Tumor Immunology , Radboud University Medical Center, Radboud Institute for Molecular Life Sciences , Nijmegen , The Netherlands
| | - Martine Bloemendal
- a Department of Medical Oncology , Radboud University Medical Center , Nijmegen , The Netherlands.,b Department of Tumor Immunology , Radboud University Medical Center, Radboud Institute for Molecular Life Sciences , Nijmegen , The Netherlands
| | - Winald R Gerritsen
- a Department of Medical Oncology , Radboud University Medical Center , Nijmegen , The Netherlands
| | - I Jolanda M de Vries
- b Department of Tumor Immunology , Radboud University Medical Center, Radboud Institute for Molecular Life Sciences , Nijmegen , The Netherlands
| | - Gerty Schreibelt
- b Department of Tumor Immunology , Radboud University Medical Center, Radboud Institute for Molecular Life Sciences , Nijmegen , The Netherlands
| |
Collapse
|
5781
|
Emerging targets for radioprotection and radiosensitization in radiotherapy. Tumour Biol 2016; 37:11589-11609. [DOI: 10.1007/s13277-016-5117-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Accepted: 06/09/2016] [Indexed: 01/12/2023] Open
|
5782
|
Ratta R, Zappasodi R, Raggi D, Grassi P, Verzoni E, Necchi A, Di Nicola M, Salvioni R, de Braud F, Procopio G. Immunotherapy advances in uro-genital malignancies. Crit Rev Oncol Hematol 2016; 105:52-64. [PMID: 27372200 DOI: 10.1016/j.critrevonc.2016.06.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 05/27/2016] [Accepted: 06/16/2016] [Indexed: 12/27/2022] Open
Abstract
Immunotherapy for the treatment of cancer has made significant progresses over the last 20 years. Multiple efforts have been attempted to restore immune-mediated tumor elimination, leading to the development of several targeted immunotherapies. Data from recent clinical trials suggest that these agents might improve the prognosis of patients with advanced genito-urinary (GU) malignancies. Nivolumab has been the first immune checkpoint-inhibitor approved for pre-treated patients with metastatic renal cell carcinoma. Pembrolizumab and atezolizumab have shown promising results in both phase I and II trials in urothelial carcinoma. Brentuximab vedotin has demonstrated early signals of clinical activity and immunomodulatory effects in highly pre-treated patients with testicular germ cell tumors. In this review, we have summarized the major clinical achievements of immunotherapy in GU cancers, focusing on immune checkpoint blockade as well as the new immunomodulatory monoclonal antibodies (mAbs) under clinical evaluation for these malignancies.
Collapse
Affiliation(s)
- Raffaele Ratta
- Department of Medical Oncology, Fondazione IRCSS Istituto Nazionale Tumori, Milan, Italy
| | - Roberta Zappasodi
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Daniele Raggi
- Department of Medical Oncology, Fondazione IRCSS Istituto Nazionale Tumori, Milan, Italy
| | - Paolo Grassi
- Department of Medical Oncology, Fondazione IRCSS Istituto Nazionale Tumori, Milan, Italy
| | - Elena Verzoni
- Department of Medical Oncology, Fondazione IRCSS Istituto Nazionale Tumori, Milan, Italy
| | - Andrea Necchi
- Department of Medical Oncology, Fondazione IRCSS Istituto Nazionale Tumori, Milan, Italy
| | - Massimo Di Nicola
- Unit of Immunotherapy and Anticancer Innovative Therapeutics, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Roberto Salvioni
- Department of Surgery, Urology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Filippo de Braud
- Department of Medical Oncology, Fondazione IRCSS Istituto Nazionale Tumori, Milan, Italy
| | - Giuseppe Procopio
- Department of Medical Oncology, Fondazione IRCSS Istituto Nazionale Tumori, Milan, Italy.
| |
Collapse
|
5783
|
Progression-free survival landmark analysis: a critical endpoint in melanoma clinical trials. Lancet Oncol 2016; 17:1037-1039. [PMID: 27324281 DOI: 10.1016/s1470-2045(16)30017-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2016] [Revised: 03/08/2016] [Accepted: 03/15/2016] [Indexed: 11/20/2022]
|
5784
|
Deficient Mismatch Repair and the Role of Immunotherapy in Metastatic Colorectal Cancer. Curr Treat Options Oncol 2016; 17:41. [DOI: 10.1007/s11864-016-0414-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
5785
|
Boudewijns S, Koornstra RHT, Westdorp H, Schreibelt G, van den Eertwegh AJM, Geukes Foppen MH, Haanen JB, de Vries IJM, Figdor CG, Bol KF, Gerritsen WR. Ipilimumab administered to metastatic melanoma patients who progressed after dendritic cell vaccination. Oncoimmunology 2016; 5:e1201625. [PMID: 27622070 PMCID: PMC5007966 DOI: 10.1080/2162402x.2016.1201625] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 06/09/2016] [Accepted: 06/09/2016] [Indexed: 11/08/2022] Open
Abstract
Background: Ipilimumab has proven to be effective in metastatic melanoma patients. The purpose of this study was to determine the efficacy of ipilimumab in advanced melanoma patients who showed progressive disease upon experimental dendritic cell (DC) vaccination. Methods: Retrospective analysis of 48 stage IV melanoma patients treated with ipilimumab after progression upon DC vaccination earlier in their treatment. DC vaccination was given either as adjuvant treatment for stage III disease (n = 18) or for stage IV disease (n = 30). Ipilimumab (3 mg/kg) was administered every 3 weeks for up to 4 cycles. Results: Median time between progression upon DC vaccination and first gift of ipilimumab was 5.4 mo. Progression-free survival (PFS) rates for patients that received ipilimumab after adjuvant DC vaccination, and patients that received DC vaccination for stage IV melanoma, were 35% and 7% at 1 y and 35% and 3% at 2 y, while the median PFS was 2.9 mo and 3.1 mo, respectively. Median overall survival of patients pre-treated with adjuvant DC vaccination for stage III melanoma was not reached versus 8.0 mo (95% CI, 5.2–10.9) in the group pre-treated with DC vaccination for stage IV disease (HR of death, 0.36; p = 0.017). Grade 3 immune-related adverse events occurred in 19% of patients and one death (2%) was related to ipilimumab. Conclusions: Clinical responses to ipilimumab were found in a considerable number of advanced melanoma patients with progression after adjuvant DC vaccination for stage III disease, while the effect was very limited in patients who showed progression after DC vaccination for stage IV disease.
Collapse
Affiliation(s)
- Steve Boudewijns
- Department of Medical Oncology, Radboud University Medical Centre, Nijmegen, the Netherlands; Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Rutger H T Koornstra
- Department of Medical Oncology, Radboud University Medical Centre , Nijmegen, the Netherlands
| | - Harm Westdorp
- Department of Medical Oncology, Radboud University Medical Centre, Nijmegen, the Netherlands; Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Gerty Schreibelt
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen, the Netherlands
| | - Alfons J M van den Eertwegh
- Department of Medical Oncology, Cancer Center Amsterdam, VU University Medical Center , Amsterdam, the Netherlands
| | - Marnix H Geukes Foppen
- Department of Medical Oncology, Netherlands Cancer Institute , Amsterdam, the Netherlands
| | - John B Haanen
- Department of Medical Oncology, Netherlands Cancer Institute , Amsterdam, the Netherlands
| | - I Jolanda M de Vries
- Department of Medical Oncology, Radboud University Medical Centre, Nijmegen, the Netherlands; Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Carl G Figdor
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center , Nijmegen, the Netherlands
| | - Kalijn F Bol
- Department of Medical Oncology, Radboud University Medical Centre, Nijmegen, the Netherlands; Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Winald R Gerritsen
- Department of Medical Oncology, Radboud University Medical Centre , Nijmegen, the Netherlands
| |
Collapse
|
5786
|
Chen PL, Roh W, Reuben A, Cooper ZA, Spencer CN, Prieto PA, Miller JP, Bassett RL, Gopalakrishnan V, Wani K, De Macedo MP, Austin-Breneman JL, Jiang H, Chang Q, Reddy SM, Chen WS, Tetzlaff MT, Broaddus RJ, Davies MA, Gershenwald JE, Haydu L, Lazar AJ, Patel SP, Hwu P, Hwu WJ, Diab A, Glitza IC, Woodman SE, Vence LM, Wistuba II, Amaria RN, Kwong LN, Prieto V, Davis RE, Ma W, Overwijk WW, Sharpe AH, Hu J, Futreal PA, Blando J, Sharma P, Allison JP, Chin L, Wargo JA. Analysis of Immune Signatures in Longitudinal Tumor Samples Yields Insight into Biomarkers of Response and Mechanisms of Resistance to Immune Checkpoint Blockade. Cancer Discov 2016; 6:827-37. [PMID: 27301722 DOI: 10.1158/2159-8290.cd-15-1545] [Citation(s) in RCA: 759] [Impact Index Per Article: 84.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Accepted: 06/10/2016] [Indexed: 11/16/2022]
Abstract
UNLABELLED Immune checkpoint blockade represents a major breakthrough in cancer therapy; however, responses are not universal. Genomic and immune features in pretreatment tumor biopsies have been reported to correlate with response in patients with melanoma and other cancers, but robust biomarkers have not been identified. We studied a cohort of patients with metastatic melanoma initially treated with cytotoxic T-lymphocyte-associated antigen-4 (CTLA4) blockade (n = 53) followed by programmed death-1 (PD-1) blockade at progression (n = 46), and analyzed immune signatures in longitudinal tissue samples collected at multiple time points during therapy. In this study, we demonstrate that adaptive immune signatures in tumor biopsy samples obtained early during the course of treatment are highly predictive of response to immune checkpoint blockade and also demonstrate differential effects on the tumor microenvironment induced by CTLA4 and PD-1 blockade. Importantly, potential mechanisms of therapeutic resistance to immune checkpoint blockade were also identified. SIGNIFICANCE These studies demonstrate that adaptive immune signatures in early on-treatment tumor biopsies are predictive of response to checkpoint blockade and yield insight into mechanisms of therapeutic resistance. These concepts have far-reaching implications in this age of precision medicine and should be explored in immune checkpoint blockade treatment across cancer types. Cancer Discov; 6(8); 827-37. ©2016 AACR.See related commentary by Teng et al., p. 818This article is highlighted in the In This Issue feature, p. 803.
Collapse
Affiliation(s)
- Pei-Ling Chen
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas. Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Whijae Roh
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Alexandre Reuben
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Zachary A Cooper
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas. Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Christine N Spencer
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Peter A Prieto
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - John P Miller
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Roland L Bassett
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Khalida Wani
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mariana Petaccia De Macedo
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jacob L Austin-Breneman
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Hong Jiang
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Qing Chang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sangeetha M Reddy
- Department of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Wei-Shen Chen
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas. Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Michael T Tetzlaff
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Russell J Broaddus
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Michael A Davies
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jeffrey E Gershenwald
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Lauren Haydu
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Alexander J Lazar
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas. Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sapna P Patel
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Patrick Hwu
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Wen-Jen Hwu
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Adi Diab
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Isabella C Glitza
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Scott E Woodman
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Luis M Vence
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ignacio I Wistuba
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Rodabe N Amaria
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Lawrence N Kwong
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Victor Prieto
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - R Eric Davis
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Wencai Ma
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Willem W Overwijk
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Arlene H Sharpe
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts
| | - Jianhua Hu
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - P Andrew Futreal
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jorge Blando
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Padmanee Sharma
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas. Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - James P Allison
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Lynda Chin
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jennifer A Wargo
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas. Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
5787
|
Lieberman NAP, Moyes KW, Crane CA. Developing immunotherapeutic strategies to target brain tumors. Expert Rev Anticancer Ther 2016; 16:775-88. [PMID: 27253692 DOI: 10.1080/14737140.2016.1192470] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
INTRODUCTION Recent years have seen rapid growth in cancer treatments that enhance the anti-tumor activities of the immune system. Collectively known as immunotherapy, modulation of the immune system has shown success treating some hematological malignancies, but has yet to be successfully applied to the treatment of patients with brain tumors. AREAS COVERED This review highlights mechanistic insights from murine studies and compiled recent clinical trial data, focusing on the most aggressive brain tumor, glioblastoma (GBM). The field has recently accumulated a critical mass of data, and we discuss past treatment failures in the context of newly developed approaches now entering clinical trials. This article provides an overview of the immunotherapeutic armamentarium currently in development for the treatment of patients with GBM, who are in dire need of safe and effective therapies. Expert commentary: Themes that emerge include the importance of mitigating the effects of an immunosuppressive tumor microenvironment and the potential for innate immune cell activation to enhance cytotoxic anti-tumor activity. Consideration of these studies as a collective may inform the design of new immunotherapies, as well as the immune monitoring protocols for patients participating in clinical trials.
Collapse
Affiliation(s)
- Nicole A P Lieberman
- a Seattle Children's Research Institute, Ben Towne Center for Childhood Cancer Research , Seattle , WA , USA
| | - Kara White Moyes
- a Seattle Children's Research Institute, Ben Towne Center for Childhood Cancer Research , Seattle , WA , USA
| | - Courtney A Crane
- a Seattle Children's Research Institute, Ben Towne Center for Childhood Cancer Research , Seattle , WA , USA.,b Department of Neurological Surgery , University of Washington School of Medicine , Seattle , WA , USA
| |
Collapse
|
5788
|
Sun WY, Lee YK, Koo JS. Expression of PD-L1 in triple-negative breast cancer based on different immunohistochemical antibodies. J Transl Med 2016; 14:173. [PMID: 27286842 PMCID: PMC4902914 DOI: 10.1186/s12967-016-0925-6] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 05/30/2016] [Indexed: 12/21/2022] Open
Abstract
Background To date, there are no effective therapeutic targeting agents for triple-negative breast cancer (TNBC), and PD-L1 has presented potential as an effective marker of immunotherapeutic agents. The aim of this study was to evaluate the expression of PD-L1 by three different immunohistochemical antibodies in TNBC. Methods Interpretation of all three PD-L1 antibodies showed good concordance among three readers (kappa value >0.610) in both cancer cells and immune cells. Using a tissue microarray (TMA) constructed from 218 cases of TNBC, we performed immunohistochemical staining using three of the most popular commercially used PD-L1 monoclonal antibodies (clones 28-8, E1L3N and SP142) in cancer cells and immune cells. Results Using various cut-off values of previous studies (1, 5, 10 and 50 %), the expression rates in cancer cells were: PD-L1 (E1L3N) (14.7, 14.7, 11.0, 2.3 %), PD-L1 (28-8) (13.3, 12.4, 10.1, 1.8 %), and PD-L1 (SP142) (11.5, 11.0, 6.9, 0.5 %), respectively. At the 5 % cut-off value, the discordance rate among the three antibodies was 6.0–10.6 % and was highest between PD-L1 (SP142) and the other two antibodies. The expression rates in immune cells were PD-L1 (E1L3N) (37.6 %), PD-L1 (28-8) (36.7 %), and PD-L1 (SP142) (19.3 %), and the discordance rate among the three antibodies ranged from 13.8 to 24.8 % and was also highest between PD-L1 (SP142) and the other two antibodies. Among stromal histologic types, higher PD-L1 expression in cancer cells and immune cells was measured in inflammatory-type (p < 0.05). The absence of PD-L1 (28-8) staining in immune cells was associated with shorter disease free survival (DFS) and overall survival (OS) (p = 0.043, and p = 0.021) in univariate analyses, and with shorter OS in multivariate Cox analysis (hazard ratio: 5.429, 95 % CI 1.214–24.28, p = 0.027). Conclusions PD-L1 detection in cancer cells and immune cells varied by antibody clone. The greatest amount of staining occurred with PD-L1 (E1L3N), followed by PD-L1 (28-8) and PD-L1 (SP142). The concordance rate among monoclonal PD-L1 antibodies was higher between PD-L1 (28-8) and PD-L1 (E1L3N). To determine the gold standard antibody and the most appropriate cut-off value, further study of the clinical trial group treated with PD-L1 inhibitor is necessary. Electronic supplementary material The online version of this article (doi:10.1186/s12967-016-0925-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Woo Young Sun
- Department of Surgery, Daejeon St. Mary's Hospital, The Catholic University of Korea College of Medicine, Seoul, South Korea
| | - Yu Kyung Lee
- Department of Pathology, Yonsei University College of Medicine, Severance Hospital, 50 Yonsei-ro, Seodaemun-gu, Seoul, 120-752, South Korea
| | - Ja Seung Koo
- Department of Pathology, Yonsei University College of Medicine, Severance Hospital, 50 Yonsei-ro, Seodaemun-gu, Seoul, 120-752, South Korea.
| |
Collapse
|
5789
|
Alhusseini M, Samantray J. Autoimmune diabetes superimposed on type 2 diabetes in a patient initiated on immunotherapy for lung cancer. DIABETES & METABOLISM 2016; 43:86-88. [PMID: 27291328 DOI: 10.1016/j.diabet.2016.05.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [MESH Headings] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 05/11/2016] [Accepted: 05/14/2016] [Indexed: 11/19/2022]
Affiliation(s)
- M Alhusseini
- Department of Internal Medicine, Division of Endocrinology, Detroit Medical Center, Wayne State University School of Medicine, 4201 St. Antoine, University Health Center, Suite 4H, 48201 Detroit, MI, USA.
| | - J Samantray
- Department of Internal Medicine, Division of Endocrinology, Detroit Medical Center, Wayne State University School of Medicine, 4201 St. Antoine, University Health Center, Suite 4H, 48201 Detroit, MI, USA
| |
Collapse
|
5790
|
Qian JM, Yu JB, Kluger HM, Chiang VLS. Timing and type of immune checkpoint therapy affect the early radiographic response of melanoma brain metastases to stereotactic radiosurgery. Cancer 2016; 122:3051-8. [PMID: 27285122 DOI: 10.1002/cncr.30138] [Citation(s) in RCA: 158] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 05/09/2016] [Accepted: 05/23/2016] [Indexed: 12/26/2022]
Abstract
BACKGROUND Growing evidence suggests that immunotherapy and radiation therapy can be synergistic in the treatment of cancer. This study was performed to determine the effect of the relative timing and type of immune checkpoint therapy on the response of melanoma brain metastases (BrMets) to treatment with stereotactic radiosurgery (SRS). METHODS Seventy-five melanoma patients with 566 BrMets were treated with both SRS and immune checkpoint therapy between 2007 and 2015 at a single institution. Immunotherapy and radiosurgery treatment of any single lesion were considered concurrent if SRS was administered within 4 weeks of immunotherapy. The impact of the timing and type of immunotherapy on the lesional response was determined with the Wilcoxon rank-sum test, which was used to compare the median percent lesion volume change 1.5, 3, and 6 months after SRS treatment, with significance determined by P = .0167 according to the Bonferroni correction for multiple comparisons. RESULTS Concurrent use of immunotherapy and SRS resulted in a significantly greater median percent reduction in the lesion volume at 1.5 (-63.1% vs -43.2%, P < .0001), 3 (-83.0% vs -52.8%, P < .0001), and 6 months (-94.9% vs -66.2%, P < .0001) in comparison with nonconcurrent therapy. The median percent reduction in the lesion volume was also significantly greater for anti-programmed cell death protein 1 (anti-PD-1) than anti-cytotoxic T-lymphocyte-associated protein 4 (anti-CTLA-4) at 1.5 (-71.1% vs -48.2%, P < .0001), 3 (-89.3% vs -66.2%, P < .0001), and 6 months (-95.1% vs -75.9%, P = .0004). CONCLUSIONS The administration of immunotherapy within 4 weeks of SRS results in an improved lesional response of melanoma BrMets in comparison with treatment separated by longer than 4 weeks. Anti-PD-1 therapy also results in a greater lesional response than anti-CTLA-4 after SRS. Cancer 2016;122:3051-3058. © 2016 American Cancer Society.
Collapse
Affiliation(s)
- Jack M Qian
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, Connecticut
| | - James B Yu
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, Connecticut
| | - Harriet M Kluger
- Department of Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Veronica L S Chiang
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, Connecticut. .,Department of Neurosurgery, Yale School of Medicine, New Haven, Connecticut.
| |
Collapse
|
5791
|
Abstract
To provide appropriate therapy and follow-up to patients with malignant melanoma, proper diagnostics are of critical importance. Targeted therapy of advanced melanoma is based on the molecular genetic analyses of tumor tissue. In addition, sequencing of genes and other genetic approaches can provide insight into the origin of melanocytic tumors and can aid in distinguishing benign from malignant lesions. In this regard, spizoid neoplasms remain a challenging entity. Aside from genetic analyses of tumor tissue, immunohistochemistry remains an essential tool in melanoma diagnostics and TNM classification. With new immunotherapies being approved for advanced melanoma, immunohistochemistry to determine PD-L1 expression has gained clinical interest. While PD-L1 expression is associated with response to PD-1 blockade, a substantial number of patients without PD-L1 expression can still experience tumor remission upon treatment. In this review, current and future developments in melanoma diagnostics with regard to molecular genetics and immunohistochemistry are summarized. The utilization of such analyses in clinical decision making is also discussed.
Collapse
Affiliation(s)
- B Schilling
- Klinik für Dermatologie, Venerologie und Allergologie, Universitätsklinikum Essen, Universität Duisburg-Essen, Partnerstandort des Deutschen Konsortium für Translationale Krebsforschung (DKTK), Hufelandstr. 55, 45147, Essen, Deutschland.
| | - K G Griewank
- Klinik für Dermatologie, Venerologie und Allergologie, Universitätsklinikum Essen, Universität Duisburg-Essen, Partnerstandort des Deutschen Konsortium für Translationale Krebsforschung (DKTK), Hufelandstr. 55, 45147, Essen, Deutschland.,Dermatopathologie bei Mainz, Bahnhofstr. 2B, 55268, Nieder-Olm, Deutschland
| |
Collapse
|
5792
|
Naidoo J, Li BT, Schindler K, Page DB. What does the future hold for immunotherapy in cancer? ANNALS OF TRANSLATIONAL MEDICINE 2016; 4:177. [PMID: 27275490 DOI: 10.21037/atm.2016.04.05] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- Jarushka Naidoo
- 1 Upper Aerodigestive Division, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD 21224, USA ; 2 Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA ; 3 Department of Dermatology, Division of General Dermatology and Dermatooncology, Medical University of Vienna, Vienna, Austria ; 4 Providence Portland Medical Center, Earl A. Chiles Research Institute, Portland, OR 97213, USA
| | - Bob T Li
- 1 Upper Aerodigestive Division, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD 21224, USA ; 2 Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA ; 3 Department of Dermatology, Division of General Dermatology and Dermatooncology, Medical University of Vienna, Vienna, Austria ; 4 Providence Portland Medical Center, Earl A. Chiles Research Institute, Portland, OR 97213, USA
| | - Katja Schindler
- 1 Upper Aerodigestive Division, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD 21224, USA ; 2 Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA ; 3 Department of Dermatology, Division of General Dermatology and Dermatooncology, Medical University of Vienna, Vienna, Austria ; 4 Providence Portland Medical Center, Earl A. Chiles Research Institute, Portland, OR 97213, USA
| | - David B Page
- 1 Upper Aerodigestive Division, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD 21224, USA ; 2 Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA ; 3 Department of Dermatology, Division of General Dermatology and Dermatooncology, Medical University of Vienna, Vienna, Austria ; 4 Providence Portland Medical Center, Earl A. Chiles Research Institute, Portland, OR 97213, USA
| |
Collapse
|
5793
|
Ni M, Hoffmann JM, Schmitt M, Schmitt A. Progress of dendritic cell-based cancer vaccines for patients with hematological malignancies. Expert Opin Biol Ther 2016; 16:1113-23. [PMID: 27238400 DOI: 10.1080/14712598.2016.1196181] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
INTRODUCTION Dendritic cells (DCs) are the most professional antigen-presenting cells eliciting cellular and humoral immune responses against cancer cells by expressing these antigens on MHC class I/II complexes to T cells. Therefore, they have been employed in many clinical trials as cancer vaccines for patients with cancer. This review focuses on the use of DCs in leukemia patients expressing leukemia-associated antigens (LAAs). AREAS COVERED The contribution of both stimulating vs. tolerogenic DCs as well as of other factors to the milieu of anti-leukemia immune responses are discussed. Several DC vaccination strategies like leukemia lysate, proteins and peptides have been developed. Next generation DC vaccines comprise transduction of DCs with retroviral vectors encoding for LAAs, cytokines and costimulatory molecules as well as transfection of DCs with naked RNA encoding for LAAs. Published as well as ongoing clinical trials are reported and critically reviewed. EXPERT OPINION Future results will demonstrate whether next-generation DCs are really superior to conventional pulsing with peptide, protein or tumor lysate. However, currently available methods based on nucleic acid transfection/transduction are tempting in terms of material production costs and time for clinical application according to good manufacturing practice (GMP).
Collapse
Affiliation(s)
- Ming Ni
- a Cellular Immunotherapy, GMP Core Facility, Department of Internal Medicine V , Heidelberg University Hospital , Heidelberg , Germany
| | - Jean-Marc Hoffmann
- a Cellular Immunotherapy, GMP Core Facility, Department of Internal Medicine V , Heidelberg University Hospital , Heidelberg , Germany
| | - Michael Schmitt
- a Cellular Immunotherapy, GMP Core Facility, Department of Internal Medicine V , Heidelberg University Hospital , Heidelberg , Germany
| | - Anita Schmitt
- a Cellular Immunotherapy, GMP Core Facility, Department of Internal Medicine V , Heidelberg University Hospital , Heidelberg , Germany
| |
Collapse
|
5794
|
Abstract
INTRODUCTION Since 2010 multiple targeted therapies and immunotherapies have been approved for the treatment of advanced melanoma. Pembrolizumab, a humanized monoclonal antibody directed against programed death receptor 1 has shown significant activity in advanced melanoma resulting in its approval first as post-ipilimumab and subsequently as frontline treatment. AREAS COVERED This article reviews the approved agents for the treatment of advanced melanoma with a focus on the preclinical and clinical evidence for the use of pembrolizumab in this setting. Primary emphasis is given to the clinical development of pembrolizumab, including phase I-III trials. Finally, we explore the role of pembrolizumab in combination with other therapies and ongoing investigations into its effectiveness in expanded patient populations. EXPERT OPINION Pembrolizumab provides durable responses and represents a major advancement in the treatment options for patients with advanced melanoma. Early studies of pembrolizumab in combination with other therapeutic agents have generated significant interest and further investigations including advanced clinical trials are warranted to evaluate safety and potential improved outcomes. Pembrolizumab and other immune checkpoint inhibitors are likely to play an expanded role in the treatment of advanced melanoma and other solid tumors over the next decade.
Collapse
Affiliation(s)
- Michael C Burns
- Vanderbilt University School of Medicine, Department of Medicine, Nashville, TN, USA
| | - Aidan O'Donnell
- Vanderbilt University School of Medicine, Department of Medicine, Nashville, TN, USA
| | - Igor Puzanov
- Vanderbilt-Ingram Cancer Center; Vanderbilt University Medical Center, Division of Hematology-Oncology, Nashville, TN, USA
| |
Collapse
|
5795
|
Serra-Bellver P, Valpione S, Lorigan P. Sequential immunotherapy regimens-expect the unexpected. Lancet Oncol 2016; 17:854-855. [PMID: 27269742 DOI: 10.1016/s1470-2045(16)30198-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 05/19/2016] [Indexed: 01/22/2023]
Affiliation(s)
| | - Sara Valpione
- Christie NHS Foundation Trust, Manchester M20 4BX UK
| | - Paul Lorigan
- Christie NHS Foundation Trust, Manchester M20 4BX UK; Institute of Cancer Sciences, University of Manchester, Manchester, UK.
| |
Collapse
|
5796
|
Weber JS, Gibney G, Sullivan RJ, Sosman JA, Slingluff CL, Lawrence DP, Logan TF, Schuchter LM, Nair S, Fecher L, Buchbinder EI, Berghorn E, Ruisi M, Kong G, Jiang J, Horak C, Hodi FS. Sequential administration of nivolumab and ipilimumab with a planned switch in patients with advanced melanoma (CheckMate 064): an open-label, randomised, phase 2 trial. Lancet Oncol 2016; 17:943-955. [PMID: 27269740 DOI: 10.1016/s1470-2045(16)30126-7] [Citation(s) in RCA: 267] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 04/28/2016] [Accepted: 04/29/2016] [Indexed: 12/20/2022]
Abstract
BACKGROUND Concurrent administration of the immune checkpoint inhibitors nivolumab and ipilimumab has shown greater efficacy than either agent alone in patients with advanced melanoma, albeit with more high-grade adverse events. We assessed whether sequential administration of nivolumab followed by ipilimumab, or the reverse sequence, could improve safety without compromising efficacy. METHODS We did this randomised, open-label, phase 2 study at nine academic medical centres in the USA. Eligible patients (aged ≥18 years) with unresectable stage III or IV melanoma (treatment-naive or who had progressed after no more than one previous systemic therapy, with an Eastern Cooperative Oncology Group performance status of 0 or 1) were randomly assigned (1:1) to induction with intravenous nivolumab 3 mg/kg every 2 weeks for six doses followed by a planned switch to intravenous ipilimumab 3 mg/kg every 3 weeks for four doses, or the reverse sequence. Randomisation was done by an independent interactive voice response system with a permuted block schedule (block size four) without stratification factors. After induction, both groups received intravenous nivolumab 3 mg/kg every 2 weeks until progression or unacceptable toxicity. The primary endpoint was treatment-related grade 3-5 adverse events until the end of the induction period (week 25), analysed in the as-treated population. Secondary endpoints were the proportion of patients who achieved a response at week 25 and disease progression at weeks 13 and 25. Overall survival was a prespecified exploratory endpoint. This study is registered with ClinicalTrials.gov, number NCT01783938, and is ongoing but no longer enrolling patients. FINDINGS Between April 30, 2013, and July 21, 2014, 140 patients were enrolled and randomly assigned to nivolumab followed by ipilimumab (n=70) or to the reverse sequence of ipilimumab followed by nivolumab (n=70), of whom 68 and 70 patients, respectively, received at least one dose of study drug and were included in the analyses. The frequencies of treatment-related grade 3-5 adverse events up to week 25 were similar in the nivolumab followed by ipilimumab group (34 [50%; 95% CI 37·6-62·4] of 68 patients) and in the ipilimumab followed by nivolumab group (30 [43%; 31·1-55·3] of 70 patients). The most common treatment-related grade 3-4 adverse events during the whole study period were colitis (ten [15%]) in the nivolumab followed by ipilimumab group vs 14 [20%] in the reverse sequence group), increased lipase (ten [15%] vs 12 [17%]), and diarrhoea (eight [12%] vs five [7%]). No treatment-related deaths occurred. The proportion of patients with a response at week 25 was higher with nivolumab followed by ipilimumab than with the reverse sequence (28 [41%; 95% CI 29·4-53·8] vs 14 [20%; 11·4-31·3]). Progression was reported in 26 (38%; 95% CI 26·7-50·8) patients in the nivolumab followed by ipilimumab group and 43 (61%; 49·0-72·8) patients in the reverse sequence group at week 13 and in 26 (38%; 26·7-50·8) and 42 (60%; 47·6-71·5) patients at week 25, respectively. After a median follow-up of 19·8 months (IQR 12·8-25·7), median overall survival was not reached in the nivolumab followed by ipilimumab group (95% CI 23·7-not reached), whereas over a median follow-up of 14·7 months (IQR 5·6-23·9) in the ipilimumab followed by nivolumab group, median overall survival was 16·9 months (95% CI 9·2-26·5; HR 0·48 [95% CI 0·29-0·80]). A higher proportion of patients in the nivolumab followed by ipilimumab group achieved 12-month overall survival than in the ipilimumab followed by nivolumab group (76%; 95% CI 64-85 vs 54%; 42-65). INTERPRETATION Nivolumab followed by ipilimumab appears to be a more clinically beneficial option compared with the reverse sequence, albeit with a higher frequency of adverse events. FUNDING Bristol-Myers Squibb.
Collapse
Affiliation(s)
- Jeffrey S Weber
- New York University Langone Medical Center, New York, NY, USA; H Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Geoff Gibney
- H Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA; Georgetown Lombardi Comprehensive Cancer Center, Washington, DC, USA
| | | | | | | | | | | | | | - Suresh Nair
- Lehigh Valley Health Network, Allentown, PA, USA
| | - Leslie Fecher
- Indiana University Simon Cancer Center, Indianapolis, IN, USA; University of Michigan, Ann Arbor, MI, USA
| | | | | | - Mary Ruisi
- Bristol-Myers Squibb, Princeton, NJ, USA
| | | | - Joel Jiang
- Bristol-Myers Squibb, Princeton, NJ, USA
| | | | | |
Collapse
|
5797
|
McNamara MJ, Hilgart-Martiszus I, Barragan Echenique DM, Linch SN, Kasiewicz MJ, Redmond WL. Interferon-γ Production by Peripheral Lymphocytes Predicts Survival of Tumor-Bearing Mice Receiving Dual PD-1/CTLA-4 Blockade. Cancer Immunol Res 2016; 4:650-7. [DOI: 10.1158/2326-6066.cir-16-0022] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Accepted: 05/06/2016] [Indexed: 11/16/2022]
|
5798
|
Zhao F, Sucker A, Horn S, Heeke C, Bielefeld N, Schrörs B, Bicker A, Lindemann M, Roesch A, Gaudernack G, Stiller M, Becker JC, Lennerz V, Wölfel T, Schadendorf D, Griewank K, Paschen A. Melanoma Lesions Independently Acquire T-cell Resistance during Metastatic Latency. Cancer Res 2016; 76:4347-58. [PMID: 27261508 DOI: 10.1158/0008-5472.can-16-0008] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 05/18/2016] [Indexed: 11/16/2022]
Abstract
Melanoma often recurs after a latency period of several years, presenting a T cell-edited phenotype that reflects a role for CD8(+) T cells in maintaining metastatic latency. Here, we report an investigation of a patient with multiple recurrent lesions, where poorly immunogenic melanoma phenotypes were found to evolve in the presence of autologous tumor antigen-specific CD8(+) T cells. Melanoma cells from two of three late recurrent metastases, developing within a 6-year latency period, lacked HLA class I expression. CD8(+) T cell-resistant, HLA class I-negative tumor cells became clinically apparent 1.5 and 6 years into stage IV disease. Genome profiling by SNP arrays revealed that HLA class I loss in both metastases originated from a shared chromosome 15q alteration and independently acquired focal B2M gene deletions. A third HLA class I haplotype-deficient lesion developed in year 3 of stage IV disease that acquired resistance toward dominant CD8(+) T-cell clonotypes targeting stage III tumor cells. At an early stage, melanoma cells showed a dedifferentiated c-Jun(high)/MITF(low) phenotype, possibly associated with immunosuppression, which contrasted with a c-Jun(low)/MITF(high) phenotype of T cell-edited tumor cells derived from late metastases. In summary, our work shows how tumor recurrences after long-term latency evolve toward T-cell resistance by independent genetic events, as a means for immune escape and immunotherapeutic resistance. Cancer Res; 76(15); 4347-58. ©2016 AACR.
Collapse
Affiliation(s)
- Fang Zhao
- Department of Dermatology, University Hospital Essen, University Duisburg-Essen and German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Essen, Germany
| | - Antje Sucker
- Department of Dermatology, University Hospital Essen, University Duisburg-Essen and German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Essen, Germany
| | - Susanne Horn
- Department of Dermatology, University Hospital Essen, University Duisburg-Essen and German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Essen, Germany
| | - Christina Heeke
- Department of Dermatology, University Hospital Essen, University Duisburg-Essen and German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Essen, Germany
| | - Nicola Bielefeld
- Department of Dermatology, University Hospital Essen, University Duisburg-Essen and German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Essen, Germany
| | - Barbara Schrörs
- Internal Medicine III, University Cancer Center (UCT) and Research Center for Immunotherapy (FZI), University Medical Center (UMC), Johannes Gutenberg University and German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, Mainz, Germany
| | - Anne Bicker
- Institute of Molecular Genetics, Genetic Engineering Research and Consulting, Johannes Gutenberg University, Mainz, Germany
| | - Monika Lindemann
- Institute for Transfusion Medicine, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Alexander Roesch
- Department of Dermatology, University Hospital Essen, University Duisburg-Essen and German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Essen, Germany
| | - Gustav Gaudernack
- Department of Immunology, Institute for Cancer Research, Oslo University Hospital-Radiumhospitalet, Oslo, Norway
| | - Mathias Stiller
- Translational Skin Cancer Research, University Hospital Essen, University Duisburg-Essen and German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Essen, Germany
| | - Jürgen C Becker
- Translational Skin Cancer Research, University Hospital Essen, University Duisburg-Essen and German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Essen, Germany
| | - Volker Lennerz
- Internal Medicine III, University Cancer Center (UCT) and Research Center for Immunotherapy (FZI), University Medical Center (UMC), Johannes Gutenberg University and German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, Mainz, Germany
| | - Thomas Wölfel
- Internal Medicine III, University Cancer Center (UCT) and Research Center for Immunotherapy (FZI), University Medical Center (UMC), Johannes Gutenberg University and German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, Mainz, Germany
| | - Dirk Schadendorf
- Department of Dermatology, University Hospital Essen, University Duisburg-Essen and German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Essen, Germany
| | - Klaus Griewank
- Department of Dermatology, University Hospital Essen, University Duisburg-Essen and German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Essen, Germany
| | - Annette Paschen
- Department of Dermatology, University Hospital Essen, University Duisburg-Essen and German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Essen, Germany.
| |
Collapse
|
5799
|
Erickson JJ, Rogers MC, Tollefson SJ, Boyd KL, Williams JV. Multiple Inhibitory Pathways Contribute to Lung CD8+ T Cell Impairment and Protect against Immunopathology during Acute Viral Respiratory Infection. THE JOURNAL OF IMMUNOLOGY 2016; 197:233-43. [PMID: 27259857 DOI: 10.4049/jimmunol.1502115] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 05/03/2016] [Indexed: 02/06/2023]
Abstract
Viruses are frequent causes of lower respiratory infection (LRI). Programmed cell death-1 (PD-1) signaling contributes to pulmonary CD8(+) T cell (TCD8) functional impairment during acute viral LRI, but the role of TCD8 impairment in viral clearance and immunopathology is unclear. We now find that human metapneumovirus infection induces virus-specific lung TCD8 that fail to produce effector cytokines or degranulate late postinfection, with minimally increased function even in the absence of PD-1 signaling. Impaired lung TCD8 upregulated multiple inhibitory receptors, including PD-1, lymphocyte activation gene 3 (LAG-3), T cell Ig mucin 3, and 2B4. Moreover, coexpression of these receptors continued to increase even after viral clearance, with most virus-specific lung TCD8 expressing three or more inhibitory receptors on day 14 postinfection. Viral infection also increased expression of inhibitory ligands by both airway epithelial cells and APCs, further establishing an inhibitory environment. In vitro Ab blockade revealed that multiple inhibitory receptors contribute to TCD8 impairment induced by either human metapneumovirus or influenza virus infection. In vivo blockade of T cell Ig mucin 3 signaling failed to enhance TCD8 function or reduce viral titers. However, blockade of LAG-3 in PD-1-deficient mice restored TCD8 effector functions but increased lung pathology, indicating that LAG-3 mediates lung TCD8 impairment in vivo and contributes to protection from immunopathology during viral clearance. These results demonstrate that an orchestrated network of pathways modifies lung TCD8 functionality during viral LRI, with PD-1 and LAG-3 serving prominent roles. Lung TCD8 impairment may prevent immunopathology but also contributes to recurrent lung infections.
Collapse
Affiliation(s)
- John J Erickson
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Meredith C Rogers
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, TN 37232; Department of Pediatrics, University of Pittsburgh School of Medicine, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, PA 15224; and
| | - Sharon J Tollefson
- Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Kelli L Boyd
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - John V Williams
- Department of Pediatrics, University of Pittsburgh School of Medicine, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, PA 15224; and Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, TN 37232
| |
Collapse
|
5800
|
Transcutaneous iontophoretic delivery of STAT3 siRNA using layer-by-layer chitosan coated gold nanoparticles to treat melanoma. Colloids Surf B Biointerfaces 2016; 146:188-97. [PMID: 27318964 DOI: 10.1016/j.colsurfb.2016.05.076] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Revised: 05/19/2016] [Accepted: 05/26/2016] [Indexed: 01/05/2023]
Abstract
Overexpression of signal transducer and activator of transcription 3 (STAT3) protein prevents apoptosis and enhances proliferation of melanocytes. The aim of this study was to investigate the feasibility of using layer-by-layer assembled gold nanoparticles (LbL-AuNP) as a carrier for iontophoretic delivery of STAT3 siRNA to treat melanoma. Chitosan coated AuNP (AuNP-CS) were prepared by direct reduction of HAuCl4 in the presence of chitosan. The AuNP-CS were then sequentially layered with siRNA and chitosan to form AuNP-CS/siRNA/CS. STAT3 siRNA replaced with scrambled siRNA or sodium alginate were used as controls. The average particle size and zeta-potential of the prepared LbL-AuNP were 150±10nm (PDI: 0.41±0.06) and 35±6mV, respectively. In vitro studies in B16F10 murine melanoma cells showed that AuNP-CS/siRNA/CS inhibited the cell growth by 49.0±0.6% and 66.0±0.2% at 0.25nM and 0.5nM STAT3 siRNA concentration, respectively. Fluorescence microscopy and flow cytometry studies showed a time dependent cell uptake of the LbL-AuNP up to 120min. Clathrin mediated endocytosis was found to be the predominant cell uptake mechanism for LbL-AuNP. STAT3 siRNA loaded LbL-AuNP reduced the STAT3 protein expression by 47.3% in B16F10 cells. Similarly, apoptosis assay showed 29% and 44% of early and late apoptotic events, respectively after treatment with STAT3 siRNA loaded LbL-AuNP. Confocal microscope and skin cryosections showed that application of 0.47mA/cm(2) of anodal iontophoresis enhanced the skin penetration of LbL-AuNP to reach viable epidermis. In conclusion, layer-by-layer chitosan coated AuNP can be developed as a carrier for iontophoretic delivery of STAT3 siRNA to treat melanoma.
Collapse
|