4201
|
The Role of Regional Therapies for in-Transit Melanoma in the Era of Improved Systemic Options. Cancers (Basel) 2015; 7:1154-77. [PMID: 26140669 PMCID: PMC4586763 DOI: 10.3390/cancers7030830] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2015] [Revised: 06/17/2015] [Accepted: 06/24/2015] [Indexed: 12/21/2022] Open
Abstract
The incidence of melanoma has been increasing at a rapid rate, with 4%–11% of all melanoma recurrences presenting as in-transit disease. Treatments for in-transit melanoma of the extremity are varied and include surgical excision, lesional injection, regional techniques and systemic therapies. Excision to clear margins is preferred; however, in cases of widespread disease, this may not be practical. Historically, intralesional therapies were generally not curative and were often used for palliation or as adjuncts to other therapies, but recent advances in oncolytic viruses may change this paradigm. Radiation as a regional therapy can be quite locally toxic and is typically relegated to disease control and symptom relief in patients with limited treatment options. Regional therapies such as isolated limb perfusion and isolated limb infusion are older therapies, but offer the ability to treat bulky disease for curative intent with a high response rate. These techniques have their associated toxicities and can be technically challenging. Historically, systemic therapy with chemotherapies and biochemotherapies were relatively ineffective and highly toxic. With the advent of novel immunotherapeutic and targeted small molecule agents for the treatment of metastatic melanoma, the armamentarium against in-transit disease has expanded. Given the multitude of options, many different combinations and sequences of therapies can be offered to patients with in-transit extremity melanoma in the contemporary era. Reported response and survival rates of the varied treatments may offer valuable information regarding treatment decisions for patients with in-transit melanoma and provide rationale for these decisions.
Collapse
|
4202
|
Congrès de l’association américaine de recherche contre le cancer — AACR 2015. ONCOLOGIE 2015. [DOI: 10.1007/s10269-015-2535-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
4203
|
Park CO, Kupper TS. The emerging role of resident memory T cells in protective immunity and inflammatory disease. Nat Med 2015; 21:688-97. [PMID: 26121195 DOI: 10.1038/nm.3883] [Citation(s) in RCA: 416] [Impact Index Per Article: 41.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 05/19/2015] [Indexed: 02/07/2023]
Abstract
Over the past decade, it has become clear that there is an important subset of memory T cells that resides in tissues-tissue-resident memory T (TRM) cells. There is an emerging understanding that TRM cells have a role in human tissue-specific immune and inflammatory diseases. Furthermore, the nature of the molecular signals that maintain TRM cells in tissues is the subject of much investigation. In addition, whereas it is logical for TRM cells to be located in barrier tissues at interfaces with the environment, these cells have also been found in brain, kidney, joint and other non-barrier tissues in humans and mice. Given the biology and behavior of these cells, it is likely that they have a role in chronic relapsing and remitting diseases of both barrier and non-barrier tissues. In this Review we discuss recent insights into the biology of TRM cells with a particular focus on their roles in disease, both proven and putative.
Collapse
Affiliation(s)
- Chang Ook Park
- Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Thomas S Kupper
- Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
4204
|
Chen TT. Milestone Survival: A Potential Intermediate Endpoint for Immune Checkpoint Inhibitors. J Natl Cancer Inst 2015; 107:djv156. [PMID: 26113579 DOI: 10.1093/jnci/djv156] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Accepted: 05/12/2015] [Indexed: 01/22/2023] Open
Abstract
Recent advancements in cancer immunotherapies offer diverse strategies for cancer treatment. Among the most promising approaches is the blockade of immune checkpoint molecules to activate antitumor immunity. With targeted immunotherapies of new mechanisms of action come greater challenges in study design and statistical analysis, as well as the need for refining clinical trial endpoints. The long-term survival and delayed clinical effects demonstrated by these therapies could result in substantial prolongation of study duration and loss of statistical power if these key attributes are not accounted for in the study design and statistical analyses. In the Brookings Conference on Clinical Cancer Research held in Washington, DC, in November 2013, several intermediate clinical endpoints, including milestone overall survival, were proposed for the evaluation of cancer immunotherapies to take into account the possibility of delayed treatment effect and to better characterize the clinical activity profile of such agents, particularly immune checkpoint inhibitors. In this manuscript, the use of milestone survival is described as a potential efficacy endpoint for immune checkpoint inhibitors in late-stage drug development that could potentially mitigate the challenge of accelerating the drug development process when the strength of this class of agents is derived from long-term follow-up.
Collapse
Affiliation(s)
- Tai-Tsang Chen
- Department of Global Biometric Sciences, Bristol-Myers Squibb, Wallingford, CT; Department of Biostatistics, Columbia University, New York, NY.
| |
Collapse
|
4205
|
Robert C, Schachter J, Long GV, Arance A, Grob JJ, Mortier L, Daud A, Carlino MS, McNeil C, Lotem M, Larkin J, Lorigan P, Neyns B, Blank CU, Hamid O, Mateus C, Shapira-Frommer R, Kosh M, Zhou H, Ibrahim N, Ebbinghaus S, Ribas A. Pembrolizumab versus Ipilimumab in Advanced Melanoma. N Engl J Med 2015; 372:2521-32. [PMID: 25891173 DOI: 10.1056/nejmoa1503093] [Citation(s) in RCA: 4448] [Impact Index Per Article: 444.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND The immune checkpoint inhibitor ipilimumab is the standard-of-care treatment for patients with advanced melanoma. Pembrolizumab inhibits the programmed cell death 1 (PD-1) immune checkpoint and has antitumor activity in patients with advanced melanoma. METHODS In this randomized, controlled, phase 3 study, we assigned 834 patients with advanced melanoma in a 1:1:1 ratio to receive pembrolizumab (at a dose of 10 mg per kilogram of body weight) every 2 weeks or every 3 weeks or four doses of ipilimumab (at 3 mg per kilogram) every 3 weeks. Primary end points were progression-free and overall survival. RESULTS The estimated 6-month progression-free-survival rates were 47.3% for pembrolizumab every 2 weeks, 46.4% for pembrolizumab every 3 weeks, and 26.5% for ipilimumab (hazard ratio for disease progression, 0.58; P<0.001 for both pembrolizumab regimens versus ipilimumab; 95% confidence intervals [CIs], 0.46 to 0.72 and 0.47 to 0.72, respectively). Estimated 12-month survival rates were 74.1%, 68.4%, and 58.2%, respectively (hazard ratio for death for pembrolizumab every 2 weeks, 0.63; 95% CI, 0.47 to 0.83; P=0.0005; hazard ratio for pembrolizumab every 3 weeks, 0.69; 95% CI, 0.52 to 0.90; P=0.0036). The response rate was improved with pembrolizumab administered every 2 weeks (33.7%) and every 3 weeks (32.9%), as compared with ipilimumab (11.9%) (P<0.001 for both comparisons). Responses were ongoing in 89.4%, 96.7%, and 87.9% of patients, respectively, after a median follow-up of 7.9 months. Efficacy was similar in the two pembrolizumab groups. Rates of treatment-related adverse events of grade 3 to 5 severity were lower in the pembrolizumab groups (13.3% and 10.1%) than in the ipilimumab group (19.9%). CONCLUSIONS The anti-PD-1 antibody pembrolizumab prolonged progression-free survival and overall survival and had less high-grade toxicity than did ipilimumab in patients with advanced melanoma. (Funded by Merck Sharp & Dohme; KEYNOTE-006 ClinicalTrials.gov number, NCT01866319.).
Collapse
|
4206
|
Waidmann O, Trojan J. Novel drugs in clinical development for hepatocellular carcinoma. Expert Opin Investig Drugs 2015; 24:1075-82. [PMID: 26108356 DOI: 10.1517/13543784.2015.1058776] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Sorafenib is the only systemic drug approved for the treatment of advanced hepatocellular carcinoma (HCC). Within recent years, several investigational agents mainly targeting angiogenesis failed in late-phase clinical development either due to toxicity or lack of benefit. AREAS COVERED This review covers recent clinical data on systemic agents and ongoing trials in patients with advanced HCC. EXPERT OPINION In unselected patients with advanced HCC, disappointing results have been reported from several large trials. However, in two subgroups encouraging results have been achieved. Treatment with the MET inhibitor tivantinib resulted in a substantial survival benefit in the subgroup of MET overexpressing tumors in a randomized Phase II trial. Furthermore, the vascular endothelial growth factor receptor 2 antibody ramucirumab resulted in improved overall survival in patients with baseline α-fetoprotein (AFP) ≥ 400 ng/ml in a Phase III trial. These two agents, and several others, will be further developed in HCC. Moreover, immunotherapeutics such as checkpoint inhibitors, programmed death receptor-1 blocking antibodies and oncolytic viruses are under investigation in advanced HCC.
Collapse
Affiliation(s)
- Oliver Waidmann
- Universitätsklinikum Frankfurt, Medizinische Klinik 1 , Theodor-Stern-Kai 7, Frankfurt am Main , Germany +49 0 69 6301 7860 ; +49 0 69 6301 6448 ;
| | | |
Collapse
|
4207
|
Schreiner J, Thommen DS, Herzig P, Bacac M, Klein C, Roller A, Belousov A, Levitsky V, Savic S, Moersig W, Uhlenbrock F, Heinzelmann-Schwarz VA, Umana P, Pisa P, von Bergwelt-Baildon M, Lardinois D, Müller P, Karanikas V, Zippelius A. Expression of inhibitory receptors on intratumoral T cells modulates the activity of a T cell-bispecific antibody targeting folate receptor. Oncoimmunology 2015; 5:e1062969. [PMID: 27057429 DOI: 10.1080/2162402x.2015.1062969] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Revised: 06/08/2015] [Accepted: 06/10/2015] [Indexed: 12/18/2022] Open
Abstract
T-cell bispecific antibodies (TCBs) are a novel therapeutic tool designed to selectively recruit T-cells to tumor cells and simultaneously activate them. However, it is currently unknown whether the dysfunctional state of T-cells, embedded into the tumor microenvironment, imprints on the therapeutic activity of TCBs. We performed a comprehensive analysis of activation and effector functions of tumor-infiltrating T-cells (TILs) in different tumor types, upon stimulation by a TCB targeting folate receptor 1 and CD3 (FolR1-TCB). We observed a considerable heterogeneity in T-cell activation, cytokine production and tumor cell killing upon exposure to FolR1-TCB among different FolR1-expressing tumors. Of note, tumors presenting with a high frequency of PD-1hi TILs displayed significantly impaired tumor cell killing and T-cell function. Further characterization of additional T-cell inhibitory receptors revealed that PD-1hi TILs defined a T-cell subset with particularly high levels of multiple inhibitory receptors compared with PD-1int and PD-1neg T-cells. PD-1 blockade could restore cytokine secretion but not cytotoxicity of TILs in a subset of patients with scarce PD-1hi expressing cells; in contrast, patients with abundance of PD-1hi expressing T-cells did not benefit from PD-1 blockade. Our data highlight that FolR1-TCB is a promising novel immunotherapeutic treatment option which is capable of activating intratumoral T-cells in different carcinomas. However, its therapeutic efficacy may be substantially hampered by a pre-existing dysfunctional state of T-cells, reflected by abundance of intratumoral PD-1hi T-cells. These findings present a rationale for combinatorial approaches of TCBs with other therapeutic strategies targeting T-cell dysfunction.
Collapse
Affiliation(s)
- Jens Schreiner
- Laboratory of Cancer Immunology, Department of Biomedicine , Basel, Switzerland
| | - Daniela S Thommen
- Laboratory of Cancer Immunology, Department of Biomedicine, Basel, Switzerland; Department of Medical Oncology, University Hospital Basel, Basel, Switzerland
| | - Petra Herzig
- Laboratory of Cancer Immunology, Department of Biomedicine , Basel, Switzerland
| | - Marina Bacac
- Roche Pharma Research and Early Development, Roche Innovation Center Zurich , Schlieren, Switzerland
| | - Christian Klein
- Roche Pharma Research and Early Development, Roche Innovation Center Zurich , Schlieren, Switzerland
| | - Andreas Roller
- Roche Pharma Research and Early Development, Roche Innovation Center Penzberg, Roche Innovation Center Penzberg , Penzberg, Germany
| | - Anton Belousov
- Roche Pharma Research and Early Development, Roche Innovation Center Penzberg, Roche Innovation Center Penzberg , Penzberg, Germany
| | - Victor Levitsky
- Roche Pharma Research and Early Development, Roche Innovation Center Zurich , Schlieren, Switzerland
| | - Spasenija Savic
- Institute of Pathology, University Hospital Basel , Basel, Switzerland
| | - Wolfgang Moersig
- Department of Surgery, University Hospital Basel , Basel, Switzerland
| | | | | | - Pablo Umana
- Roche Pharma Research and Early Development, Roche Innovation Center Zurich , Schlieren, Switzerland
| | - Pavel Pisa
- Roche Pharma Research and Early Development, Roche Innovation Center Zurich , Schlieren, Switzerland
| | | | - Didier Lardinois
- Department of Surgery, University Hospital Basel , Basel, Switzerland
| | - Philipp Müller
- Laboratory of Cancer Immunology, Department of Biomedicine , Basel, Switzerland
| | - Vaios Karanikas
- Roche Pharma Research and Early Development, Roche Innovation Center Zurich , Schlieren, Switzerland
| | - Alfred Zippelius
- Laboratory of Cancer Immunology, Department of Biomedicine, Basel, Switzerland; Department of Medical Oncology, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
4208
|
Ribas A, Puzanov I, Dummer R, Schadendorf D, Hamid O, Robert C, Hodi FS, Schachter J, Pavlick AC, Lewis KD, Cranmer LD, Blank CU, O'Day SJ, Ascierto PA, Salama AKS, Margolin KA, Loquai C, Eigentler TK, Gangadhar TC, Carlino MS, Agarwala SS, Moschos SJ, Sosman JA, Goldinger SM, Shapira-Frommer R, Gonzalez R, Kirkwood JM, Wolchok JD, Eggermont A, Li XN, Zhou W, Zernhelt AM, Lis J, Ebbinghaus S, Kang SP, Daud A. Pembrolizumab versus investigator-choice chemotherapy for ipilimumab-refractory melanoma (KEYNOTE-002): a randomised, controlled, phase 2 trial. Lancet Oncol 2015; 16:908-18. [PMID: 26115796 DOI: 10.1016/s1470-2045(15)00083-2] [Citation(s) in RCA: 1260] [Impact Index Per Article: 126.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2015] [Revised: 05/15/2015] [Accepted: 05/18/2015] [Indexed: 02/08/2023]
Abstract
BACKGROUND Patients with melanoma that progresses on ipilimumab and, if BRAF(V600) mutant-positive, a BRAF or MEK inhibitor or both, have few treatment options. We assessed the efficacy and safety of two pembrolizumab doses versus investigator-choice chemotherapy in patients with ipilimumab-refractory melanoma. METHODS We carried out a randomised phase 2 trial of patients aged 18 years or older from 73 hospitals, clinics, and academic medical centres in 12 countries who had confirmed progressive disease within 24 weeks after two or more ipilimumab doses and, if BRAF(V600) mutant-positive, previous treatment with a BRAF or MEK inhibitor or both. Patients had to have resolution of all ipilimumab-related adverse events to grade 0-1 and prednisone 10 mg/day or less for at least 2 weeks, an Eastern Cooperative Oncology Group (ECOG) performance status of 0 or 1, and at least one measurable lesion to be eligible. Using a centralised interactive voice response system, we randomly assigned (1:1:1) patients in a block size of six to receive intravenous pembrolizumab 2 mg/kg or 10 mg/kg every 3 weeks or investigator-choice chemotherapy (paclitaxel plus carboplatin, paclitaxel, carboplatin, dacarbazine, or oral temozolomide). Randomisation was stratified by ECOG performance status, lactate dehydrogenase concentration, and BRAF(V600) mutation status. Individual treatment assignment between pembrolizumab and chemotherapy was open label, but investigators and patients were masked to assignment of the dose of pembrolizumab. We present the primary endpoint at the prespecified second interim analysis of progression-free survival in the intention-to-treat population. This study is registered with ClinicalTrials.gov, number NCT01704287. The study is closed to enrolment but continues to follow up and treat patients. FINDINGS Between Nov 30, 2012, and Nov 13, 2013, we enrolled 540 patients: 180 patients were randomly assigned to receive pembrolizumab 2 mg/kg, 181 to receive pembrolizumab 10 mg/kg, and 179 to receive chemotherapy. Based on 410 progression-free survival events, progression-free survival was improved in patients assigned to pembrolizumab 2 mg/kg (HR 0·57, 95% CI 0·45-0·73; p<0·0001) and those assigned to pembrolizumab 10 mg/kg (0·50, 0·39-0·64; p<0·0001) compared with those assigned to chemotherapy. 6-month progression-free survival was 34% (95% CI 27-41) in the pembrolizumab 2 mg/kg group, 38% (31-45) in the 10 mg/kg group, and 16% (10-22) in the chemotherapy group. Treatment-related grade 3-4 adverse events occurred in 20 (11%) patients in the pembrolizumab 2 mg/kg group, 25 (14%) in the pembrolizumab 10 mg/kg group, and 45 (26%) in the chemotherapy group. The most common treatment-related grade 3-4 adverse event in the pembrolizumab groups was fatigue (two [1%] of 178 patients in the 2 mg/kg group and one [<1%] of 179 patients in the 10 mg/kg group, compared with eight [5%] of 171 in the chemotherapy group). Other treatment-related grade 3-4 adverse events include generalised oedema and myalgia (each in two [1%] patients) in those given pembrolizumab 2 mg/kg; hypopituitarism, colitis, diarrhoea, decreased appetite, hyponatremia, and pneumonitis (each in two [1%]) in those given pembrolizumab 10 mg/kg; and anaemia (nine [5%]), fatigue (eight [5%]), neutropenia (six [4%]), and leucopenia (six [4%]) in those assigned to chemotherapy. INTERPRETATION These findings establish pembrolizumab as a new standard of care for the treatment of ipilimumab-refractory melanoma. FUNDING Merck Sharp & Dohme.
Collapse
Affiliation(s)
- Antoni Ribas
- University of California, Los Angeles, Los Angeles, CA, USA.
| | - Igor Puzanov
- Vanderbilt-Ingram Cancer Center, Nashville, TN, USA
| | | | | | - Omid Hamid
- The Angeles Clinic and Research Institute, Los Angeles, CA, USA
| | | | | | | | | | | | - Lee D Cranmer
- University of Arizona Cancer Center, Tucson, AZ, USA
| | | | | | | | | | - Kim A Margolin
- Seattle Cancer Care Alliance/University of Washington, Seattle, WA, USA
| | | | | | - Tara C Gangadhar
- Abramson Cancer Center of the University of Pennsylvania, Philadelphia, PA, USA
| | - Matteo S Carlino
- Crown Princess Mary Cancer Centre, Westmead and Blacktown Hospitals, and Melanoma Institute Australia, Westmead, NSW, Australia
| | - Sanjiv S Agarwala
- St Luke's Cancer Center, Bethlehem, PA, USA; Temple University, Philadelphia, PA, USA
| | | | | | | | | | | | | | - Jedd D Wolchok
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | | | | | | | - Joy Lis
- Merck & Co, Kenilworth, NJ, USA
| | | | | | - Adil Daud
- University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
4209
|
Hauschild A, Garbe C. Immunotherapy: Combined immunotherapy--a new standard in metastatic melanoma? Nat Rev Clin Oncol 2015; 12:439-40. [PMID: 26099985 DOI: 10.1038/nrclinonc.2015.118] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Axel Hauschild
- University Hospital of Schleswig-Holstein, Campus Kiel, Department of Dermatology, Schittenhelmstrasse 7, 24105 Kiel, Germany
| | - Claus Garbe
- Division of Dermatooncology, Department of Dermatology, University Medical Center, Liebermeisterstrasse 25, 72074 Tuebingen, Germany
| |
Collapse
|
4210
|
Gettinger SN, Horn L, Gandhi L, Spigel DR, Antonia SJ, Rizvi NA, Powderly JD, Heist RS, Carvajal RD, Jackman DM, Sequist LV, Smith DC, Leming P, Carbone DP, Pinder-Schenck MC, Topalian SL, Hodi FS, Sosman JA, Sznol M, McDermott DF, Pardoll DM, Sankar V, Ahlers CM, Salvati M, Wigginton JM, Hellmann MD, Kollia GD, Gupta AK, Brahmer JR. Overall Survival and Long-Term Safety of Nivolumab (Anti-Programmed Death 1 Antibody, BMS-936558, ONO-4538) in Patients With Previously Treated Advanced Non-Small-Cell Lung Cancer. J Clin Oncol 2015; 33:2004-12. [PMID: 25897158 PMCID: PMC4672027 DOI: 10.1200/jco.2014.58.3708] [Citation(s) in RCA: 953] [Impact Index Per Article: 95.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE Programmed death 1 is an immune checkpoint that suppresses antitumor immunity. Nivolumab, a fully human immunoglobulin G4 programmed death 1 immune checkpoint inhibitor antibody, was active and generally well tolerated in patients with advanced solid tumors treated in a phase I trial with expansion cohorts. We report overall survival (OS), response durability, and long-term safety in patients with non-small-cell lung cancer (NSCLC) receiving nivolumab in this trial. PATIENTS AND METHODS Patients (N = 129) with heavily pretreated advanced NSCLC received nivolumab 1, 3, or 10 mg/kg intravenously once every 2 weeks in 8-week cycles for up to 96 weeks. Tumor burden was assessed by RECIST (version 1.0) after each cycle. RESULTS Median OS across doses was 9.9 months; 1-, 2-, and 3-year OS rates were 42%, 24%, and 18%, respectively, across doses and 56%, 42%, and 27%, respectively, at the 3-mg/kg dose (n = 37) chosen for further clinical development. Among 22 patients (17%) with objective responses, estimated median response duration was 17.0 months. An additional six patients (5%) had unconventional immune-pattern responses. Response rates were similar in squamous and nonsquamous NSCLC. Eighteen responding patients discontinued nivolumab for reasons other than progressive disease; nine (50%) of those had responses lasting > 9 months after their last dose. Grade 3 to 4 treatment-related adverse events occurred in 14% of patients. Three treatment-related deaths (2% of patients) occurred, each associated with pneumonitis. CONCLUSION Nivolumab monotherapy produced durable responses and encouraging survival rates in patients with heavily pretreated NSCLC. Randomized clinical trials with nivolumab in advanced NSCLC are ongoing.
Collapse
Affiliation(s)
- Scott N Gettinger
- Scott N. Gettinger and Mario Sznol, Yale Cancer Center, New Haven, CT; Leora Horn, David P. Carbone, and Jeffrey A. Sosman, Vanderbilt University Medical Center; David R. Spigel, Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN; Leena Gandhi, David M. Jackman, and F. Stephen Hodi, Dana-Farber Cancer Institute; Rebecca S. Heist and Lecia V. Sequist, Massachusetts General Hospital Cancer Center; David F. McDermott, Beth Israel Deaconess Medical Center, Boston, MA; Scott J. Antonia and Mary C. Pinder-Schenck, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Naiyer A. Rizvi, Richard D. Carvajal, and Matthew D. Hellmann, Memorial Sloan Kettering Cancer Center, New York, NY; John D. Powderly, Carolina BioOncology Institute, Huntersville, NC; David C. Smith, University of Michigan, Ann Arbor, MI; Philip Leming, Christ Hospital Cancer Center, Cincinnati, OH; Suzanne L. Topalian, Drew M. Pardoll, and Julie R. Brahmer, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, MD; and Vindira Sankar, Christoph M. Ahlers, Mark Salvati, Jon M. Wigginton, Georgia D. Kollia, and Ashok K. Gupta, Bristol-Myers Squibb, Princeton, NJ.
| | - Leora Horn
- Scott N. Gettinger and Mario Sznol, Yale Cancer Center, New Haven, CT; Leora Horn, David P. Carbone, and Jeffrey A. Sosman, Vanderbilt University Medical Center; David R. Spigel, Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN; Leena Gandhi, David M. Jackman, and F. Stephen Hodi, Dana-Farber Cancer Institute; Rebecca S. Heist and Lecia V. Sequist, Massachusetts General Hospital Cancer Center; David F. McDermott, Beth Israel Deaconess Medical Center, Boston, MA; Scott J. Antonia and Mary C. Pinder-Schenck, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Naiyer A. Rizvi, Richard D. Carvajal, and Matthew D. Hellmann, Memorial Sloan Kettering Cancer Center, New York, NY; John D. Powderly, Carolina BioOncology Institute, Huntersville, NC; David C. Smith, University of Michigan, Ann Arbor, MI; Philip Leming, Christ Hospital Cancer Center, Cincinnati, OH; Suzanne L. Topalian, Drew M. Pardoll, and Julie R. Brahmer, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, MD; and Vindira Sankar, Christoph M. Ahlers, Mark Salvati, Jon M. Wigginton, Georgia D. Kollia, and Ashok K. Gupta, Bristol-Myers Squibb, Princeton, NJ
| | - Leena Gandhi
- Scott N. Gettinger and Mario Sznol, Yale Cancer Center, New Haven, CT; Leora Horn, David P. Carbone, and Jeffrey A. Sosman, Vanderbilt University Medical Center; David R. Spigel, Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN; Leena Gandhi, David M. Jackman, and F. Stephen Hodi, Dana-Farber Cancer Institute; Rebecca S. Heist and Lecia V. Sequist, Massachusetts General Hospital Cancer Center; David F. McDermott, Beth Israel Deaconess Medical Center, Boston, MA; Scott J. Antonia and Mary C. Pinder-Schenck, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Naiyer A. Rizvi, Richard D. Carvajal, and Matthew D. Hellmann, Memorial Sloan Kettering Cancer Center, New York, NY; John D. Powderly, Carolina BioOncology Institute, Huntersville, NC; David C. Smith, University of Michigan, Ann Arbor, MI; Philip Leming, Christ Hospital Cancer Center, Cincinnati, OH; Suzanne L. Topalian, Drew M. Pardoll, and Julie R. Brahmer, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, MD; and Vindira Sankar, Christoph M. Ahlers, Mark Salvati, Jon M. Wigginton, Georgia D. Kollia, and Ashok K. Gupta, Bristol-Myers Squibb, Princeton, NJ
| | - David R Spigel
- Scott N. Gettinger and Mario Sznol, Yale Cancer Center, New Haven, CT; Leora Horn, David P. Carbone, and Jeffrey A. Sosman, Vanderbilt University Medical Center; David R. Spigel, Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN; Leena Gandhi, David M. Jackman, and F. Stephen Hodi, Dana-Farber Cancer Institute; Rebecca S. Heist and Lecia V. Sequist, Massachusetts General Hospital Cancer Center; David F. McDermott, Beth Israel Deaconess Medical Center, Boston, MA; Scott J. Antonia and Mary C. Pinder-Schenck, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Naiyer A. Rizvi, Richard D. Carvajal, and Matthew D. Hellmann, Memorial Sloan Kettering Cancer Center, New York, NY; John D. Powderly, Carolina BioOncology Institute, Huntersville, NC; David C. Smith, University of Michigan, Ann Arbor, MI; Philip Leming, Christ Hospital Cancer Center, Cincinnati, OH; Suzanne L. Topalian, Drew M. Pardoll, and Julie R. Brahmer, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, MD; and Vindira Sankar, Christoph M. Ahlers, Mark Salvati, Jon M. Wigginton, Georgia D. Kollia, and Ashok K. Gupta, Bristol-Myers Squibb, Princeton, NJ
| | - Scott J Antonia
- Scott N. Gettinger and Mario Sznol, Yale Cancer Center, New Haven, CT; Leora Horn, David P. Carbone, and Jeffrey A. Sosman, Vanderbilt University Medical Center; David R. Spigel, Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN; Leena Gandhi, David M. Jackman, and F. Stephen Hodi, Dana-Farber Cancer Institute; Rebecca S. Heist and Lecia V. Sequist, Massachusetts General Hospital Cancer Center; David F. McDermott, Beth Israel Deaconess Medical Center, Boston, MA; Scott J. Antonia and Mary C. Pinder-Schenck, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Naiyer A. Rizvi, Richard D. Carvajal, and Matthew D. Hellmann, Memorial Sloan Kettering Cancer Center, New York, NY; John D. Powderly, Carolina BioOncology Institute, Huntersville, NC; David C. Smith, University of Michigan, Ann Arbor, MI; Philip Leming, Christ Hospital Cancer Center, Cincinnati, OH; Suzanne L. Topalian, Drew M. Pardoll, and Julie R. Brahmer, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, MD; and Vindira Sankar, Christoph M. Ahlers, Mark Salvati, Jon M. Wigginton, Georgia D. Kollia, and Ashok K. Gupta, Bristol-Myers Squibb, Princeton, NJ
| | - Naiyer A Rizvi
- Scott N. Gettinger and Mario Sznol, Yale Cancer Center, New Haven, CT; Leora Horn, David P. Carbone, and Jeffrey A. Sosman, Vanderbilt University Medical Center; David R. Spigel, Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN; Leena Gandhi, David M. Jackman, and F. Stephen Hodi, Dana-Farber Cancer Institute; Rebecca S. Heist and Lecia V. Sequist, Massachusetts General Hospital Cancer Center; David F. McDermott, Beth Israel Deaconess Medical Center, Boston, MA; Scott J. Antonia and Mary C. Pinder-Schenck, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Naiyer A. Rizvi, Richard D. Carvajal, and Matthew D. Hellmann, Memorial Sloan Kettering Cancer Center, New York, NY; John D. Powderly, Carolina BioOncology Institute, Huntersville, NC; David C. Smith, University of Michigan, Ann Arbor, MI; Philip Leming, Christ Hospital Cancer Center, Cincinnati, OH; Suzanne L. Topalian, Drew M. Pardoll, and Julie R. Brahmer, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, MD; and Vindira Sankar, Christoph M. Ahlers, Mark Salvati, Jon M. Wigginton, Georgia D. Kollia, and Ashok K. Gupta, Bristol-Myers Squibb, Princeton, NJ
| | - John D Powderly
- Scott N. Gettinger and Mario Sznol, Yale Cancer Center, New Haven, CT; Leora Horn, David P. Carbone, and Jeffrey A. Sosman, Vanderbilt University Medical Center; David R. Spigel, Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN; Leena Gandhi, David M. Jackman, and F. Stephen Hodi, Dana-Farber Cancer Institute; Rebecca S. Heist and Lecia V. Sequist, Massachusetts General Hospital Cancer Center; David F. McDermott, Beth Israel Deaconess Medical Center, Boston, MA; Scott J. Antonia and Mary C. Pinder-Schenck, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Naiyer A. Rizvi, Richard D. Carvajal, and Matthew D. Hellmann, Memorial Sloan Kettering Cancer Center, New York, NY; John D. Powderly, Carolina BioOncology Institute, Huntersville, NC; David C. Smith, University of Michigan, Ann Arbor, MI; Philip Leming, Christ Hospital Cancer Center, Cincinnati, OH; Suzanne L. Topalian, Drew M. Pardoll, and Julie R. Brahmer, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, MD; and Vindira Sankar, Christoph M. Ahlers, Mark Salvati, Jon M. Wigginton, Georgia D. Kollia, and Ashok K. Gupta, Bristol-Myers Squibb, Princeton, NJ
| | - Rebecca S Heist
- Scott N. Gettinger and Mario Sznol, Yale Cancer Center, New Haven, CT; Leora Horn, David P. Carbone, and Jeffrey A. Sosman, Vanderbilt University Medical Center; David R. Spigel, Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN; Leena Gandhi, David M. Jackman, and F. Stephen Hodi, Dana-Farber Cancer Institute; Rebecca S. Heist and Lecia V. Sequist, Massachusetts General Hospital Cancer Center; David F. McDermott, Beth Israel Deaconess Medical Center, Boston, MA; Scott J. Antonia and Mary C. Pinder-Schenck, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Naiyer A. Rizvi, Richard D. Carvajal, and Matthew D. Hellmann, Memorial Sloan Kettering Cancer Center, New York, NY; John D. Powderly, Carolina BioOncology Institute, Huntersville, NC; David C. Smith, University of Michigan, Ann Arbor, MI; Philip Leming, Christ Hospital Cancer Center, Cincinnati, OH; Suzanne L. Topalian, Drew M. Pardoll, and Julie R. Brahmer, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, MD; and Vindira Sankar, Christoph M. Ahlers, Mark Salvati, Jon M. Wigginton, Georgia D. Kollia, and Ashok K. Gupta, Bristol-Myers Squibb, Princeton, NJ
| | - Richard D Carvajal
- Scott N. Gettinger and Mario Sznol, Yale Cancer Center, New Haven, CT; Leora Horn, David P. Carbone, and Jeffrey A. Sosman, Vanderbilt University Medical Center; David R. Spigel, Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN; Leena Gandhi, David M. Jackman, and F. Stephen Hodi, Dana-Farber Cancer Institute; Rebecca S. Heist and Lecia V. Sequist, Massachusetts General Hospital Cancer Center; David F. McDermott, Beth Israel Deaconess Medical Center, Boston, MA; Scott J. Antonia and Mary C. Pinder-Schenck, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Naiyer A. Rizvi, Richard D. Carvajal, and Matthew D. Hellmann, Memorial Sloan Kettering Cancer Center, New York, NY; John D. Powderly, Carolina BioOncology Institute, Huntersville, NC; David C. Smith, University of Michigan, Ann Arbor, MI; Philip Leming, Christ Hospital Cancer Center, Cincinnati, OH; Suzanne L. Topalian, Drew M. Pardoll, and Julie R. Brahmer, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, MD; and Vindira Sankar, Christoph M. Ahlers, Mark Salvati, Jon M. Wigginton, Georgia D. Kollia, and Ashok K. Gupta, Bristol-Myers Squibb, Princeton, NJ
| | - David M Jackman
- Scott N. Gettinger and Mario Sznol, Yale Cancer Center, New Haven, CT; Leora Horn, David P. Carbone, and Jeffrey A. Sosman, Vanderbilt University Medical Center; David R. Spigel, Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN; Leena Gandhi, David M. Jackman, and F. Stephen Hodi, Dana-Farber Cancer Institute; Rebecca S. Heist and Lecia V. Sequist, Massachusetts General Hospital Cancer Center; David F. McDermott, Beth Israel Deaconess Medical Center, Boston, MA; Scott J. Antonia and Mary C. Pinder-Schenck, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Naiyer A. Rizvi, Richard D. Carvajal, and Matthew D. Hellmann, Memorial Sloan Kettering Cancer Center, New York, NY; John D. Powderly, Carolina BioOncology Institute, Huntersville, NC; David C. Smith, University of Michigan, Ann Arbor, MI; Philip Leming, Christ Hospital Cancer Center, Cincinnati, OH; Suzanne L. Topalian, Drew M. Pardoll, and Julie R. Brahmer, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, MD; and Vindira Sankar, Christoph M. Ahlers, Mark Salvati, Jon M. Wigginton, Georgia D. Kollia, and Ashok K. Gupta, Bristol-Myers Squibb, Princeton, NJ
| | - Lecia V Sequist
- Scott N. Gettinger and Mario Sznol, Yale Cancer Center, New Haven, CT; Leora Horn, David P. Carbone, and Jeffrey A. Sosman, Vanderbilt University Medical Center; David R. Spigel, Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN; Leena Gandhi, David M. Jackman, and F. Stephen Hodi, Dana-Farber Cancer Institute; Rebecca S. Heist and Lecia V. Sequist, Massachusetts General Hospital Cancer Center; David F. McDermott, Beth Israel Deaconess Medical Center, Boston, MA; Scott J. Antonia and Mary C. Pinder-Schenck, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Naiyer A. Rizvi, Richard D. Carvajal, and Matthew D. Hellmann, Memorial Sloan Kettering Cancer Center, New York, NY; John D. Powderly, Carolina BioOncology Institute, Huntersville, NC; David C. Smith, University of Michigan, Ann Arbor, MI; Philip Leming, Christ Hospital Cancer Center, Cincinnati, OH; Suzanne L. Topalian, Drew M. Pardoll, and Julie R. Brahmer, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, MD; and Vindira Sankar, Christoph M. Ahlers, Mark Salvati, Jon M. Wigginton, Georgia D. Kollia, and Ashok K. Gupta, Bristol-Myers Squibb, Princeton, NJ
| | - David C Smith
- Scott N. Gettinger and Mario Sznol, Yale Cancer Center, New Haven, CT; Leora Horn, David P. Carbone, and Jeffrey A. Sosman, Vanderbilt University Medical Center; David R. Spigel, Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN; Leena Gandhi, David M. Jackman, and F. Stephen Hodi, Dana-Farber Cancer Institute; Rebecca S. Heist and Lecia V. Sequist, Massachusetts General Hospital Cancer Center; David F. McDermott, Beth Israel Deaconess Medical Center, Boston, MA; Scott J. Antonia and Mary C. Pinder-Schenck, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Naiyer A. Rizvi, Richard D. Carvajal, and Matthew D. Hellmann, Memorial Sloan Kettering Cancer Center, New York, NY; John D. Powderly, Carolina BioOncology Institute, Huntersville, NC; David C. Smith, University of Michigan, Ann Arbor, MI; Philip Leming, Christ Hospital Cancer Center, Cincinnati, OH; Suzanne L. Topalian, Drew M. Pardoll, and Julie R. Brahmer, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, MD; and Vindira Sankar, Christoph M. Ahlers, Mark Salvati, Jon M. Wigginton, Georgia D. Kollia, and Ashok K. Gupta, Bristol-Myers Squibb, Princeton, NJ
| | - Philip Leming
- Scott N. Gettinger and Mario Sznol, Yale Cancer Center, New Haven, CT; Leora Horn, David P. Carbone, and Jeffrey A. Sosman, Vanderbilt University Medical Center; David R. Spigel, Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN; Leena Gandhi, David M. Jackman, and F. Stephen Hodi, Dana-Farber Cancer Institute; Rebecca S. Heist and Lecia V. Sequist, Massachusetts General Hospital Cancer Center; David F. McDermott, Beth Israel Deaconess Medical Center, Boston, MA; Scott J. Antonia and Mary C. Pinder-Schenck, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Naiyer A. Rizvi, Richard D. Carvajal, and Matthew D. Hellmann, Memorial Sloan Kettering Cancer Center, New York, NY; John D. Powderly, Carolina BioOncology Institute, Huntersville, NC; David C. Smith, University of Michigan, Ann Arbor, MI; Philip Leming, Christ Hospital Cancer Center, Cincinnati, OH; Suzanne L. Topalian, Drew M. Pardoll, and Julie R. Brahmer, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, MD; and Vindira Sankar, Christoph M. Ahlers, Mark Salvati, Jon M. Wigginton, Georgia D. Kollia, and Ashok K. Gupta, Bristol-Myers Squibb, Princeton, NJ
| | - David P Carbone
- Scott N. Gettinger and Mario Sznol, Yale Cancer Center, New Haven, CT; Leora Horn, David P. Carbone, and Jeffrey A. Sosman, Vanderbilt University Medical Center; David R. Spigel, Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN; Leena Gandhi, David M. Jackman, and F. Stephen Hodi, Dana-Farber Cancer Institute; Rebecca S. Heist and Lecia V. Sequist, Massachusetts General Hospital Cancer Center; David F. McDermott, Beth Israel Deaconess Medical Center, Boston, MA; Scott J. Antonia and Mary C. Pinder-Schenck, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Naiyer A. Rizvi, Richard D. Carvajal, and Matthew D. Hellmann, Memorial Sloan Kettering Cancer Center, New York, NY; John D. Powderly, Carolina BioOncology Institute, Huntersville, NC; David C. Smith, University of Michigan, Ann Arbor, MI; Philip Leming, Christ Hospital Cancer Center, Cincinnati, OH; Suzanne L. Topalian, Drew M. Pardoll, and Julie R. Brahmer, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, MD; and Vindira Sankar, Christoph M. Ahlers, Mark Salvati, Jon M. Wigginton, Georgia D. Kollia, and Ashok K. Gupta, Bristol-Myers Squibb, Princeton, NJ
| | - Mary C Pinder-Schenck
- Scott N. Gettinger and Mario Sznol, Yale Cancer Center, New Haven, CT; Leora Horn, David P. Carbone, and Jeffrey A. Sosman, Vanderbilt University Medical Center; David R. Spigel, Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN; Leena Gandhi, David M. Jackman, and F. Stephen Hodi, Dana-Farber Cancer Institute; Rebecca S. Heist and Lecia V. Sequist, Massachusetts General Hospital Cancer Center; David F. McDermott, Beth Israel Deaconess Medical Center, Boston, MA; Scott J. Antonia and Mary C. Pinder-Schenck, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Naiyer A. Rizvi, Richard D. Carvajal, and Matthew D. Hellmann, Memorial Sloan Kettering Cancer Center, New York, NY; John D. Powderly, Carolina BioOncology Institute, Huntersville, NC; David C. Smith, University of Michigan, Ann Arbor, MI; Philip Leming, Christ Hospital Cancer Center, Cincinnati, OH; Suzanne L. Topalian, Drew M. Pardoll, and Julie R. Brahmer, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, MD; and Vindira Sankar, Christoph M. Ahlers, Mark Salvati, Jon M. Wigginton, Georgia D. Kollia, and Ashok K. Gupta, Bristol-Myers Squibb, Princeton, NJ
| | - Suzanne L Topalian
- Scott N. Gettinger and Mario Sznol, Yale Cancer Center, New Haven, CT; Leora Horn, David P. Carbone, and Jeffrey A. Sosman, Vanderbilt University Medical Center; David R. Spigel, Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN; Leena Gandhi, David M. Jackman, and F. Stephen Hodi, Dana-Farber Cancer Institute; Rebecca S. Heist and Lecia V. Sequist, Massachusetts General Hospital Cancer Center; David F. McDermott, Beth Israel Deaconess Medical Center, Boston, MA; Scott J. Antonia and Mary C. Pinder-Schenck, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Naiyer A. Rizvi, Richard D. Carvajal, and Matthew D. Hellmann, Memorial Sloan Kettering Cancer Center, New York, NY; John D. Powderly, Carolina BioOncology Institute, Huntersville, NC; David C. Smith, University of Michigan, Ann Arbor, MI; Philip Leming, Christ Hospital Cancer Center, Cincinnati, OH; Suzanne L. Topalian, Drew M. Pardoll, and Julie R. Brahmer, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, MD; and Vindira Sankar, Christoph M. Ahlers, Mark Salvati, Jon M. Wigginton, Georgia D. Kollia, and Ashok K. Gupta, Bristol-Myers Squibb, Princeton, NJ
| | - F Stephen Hodi
- Scott N. Gettinger and Mario Sznol, Yale Cancer Center, New Haven, CT; Leora Horn, David P. Carbone, and Jeffrey A. Sosman, Vanderbilt University Medical Center; David R. Spigel, Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN; Leena Gandhi, David M. Jackman, and F. Stephen Hodi, Dana-Farber Cancer Institute; Rebecca S. Heist and Lecia V. Sequist, Massachusetts General Hospital Cancer Center; David F. McDermott, Beth Israel Deaconess Medical Center, Boston, MA; Scott J. Antonia and Mary C. Pinder-Schenck, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Naiyer A. Rizvi, Richard D. Carvajal, and Matthew D. Hellmann, Memorial Sloan Kettering Cancer Center, New York, NY; John D. Powderly, Carolina BioOncology Institute, Huntersville, NC; David C. Smith, University of Michigan, Ann Arbor, MI; Philip Leming, Christ Hospital Cancer Center, Cincinnati, OH; Suzanne L. Topalian, Drew M. Pardoll, and Julie R. Brahmer, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, MD; and Vindira Sankar, Christoph M. Ahlers, Mark Salvati, Jon M. Wigginton, Georgia D. Kollia, and Ashok K. Gupta, Bristol-Myers Squibb, Princeton, NJ
| | - Jeffrey A Sosman
- Scott N. Gettinger and Mario Sznol, Yale Cancer Center, New Haven, CT; Leora Horn, David P. Carbone, and Jeffrey A. Sosman, Vanderbilt University Medical Center; David R. Spigel, Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN; Leena Gandhi, David M. Jackman, and F. Stephen Hodi, Dana-Farber Cancer Institute; Rebecca S. Heist and Lecia V. Sequist, Massachusetts General Hospital Cancer Center; David F. McDermott, Beth Israel Deaconess Medical Center, Boston, MA; Scott J. Antonia and Mary C. Pinder-Schenck, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Naiyer A. Rizvi, Richard D. Carvajal, and Matthew D. Hellmann, Memorial Sloan Kettering Cancer Center, New York, NY; John D. Powderly, Carolina BioOncology Institute, Huntersville, NC; David C. Smith, University of Michigan, Ann Arbor, MI; Philip Leming, Christ Hospital Cancer Center, Cincinnati, OH; Suzanne L. Topalian, Drew M. Pardoll, and Julie R. Brahmer, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, MD; and Vindira Sankar, Christoph M. Ahlers, Mark Salvati, Jon M. Wigginton, Georgia D. Kollia, and Ashok K. Gupta, Bristol-Myers Squibb, Princeton, NJ
| | - Mario Sznol
- Scott N. Gettinger and Mario Sznol, Yale Cancer Center, New Haven, CT; Leora Horn, David P. Carbone, and Jeffrey A. Sosman, Vanderbilt University Medical Center; David R. Spigel, Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN; Leena Gandhi, David M. Jackman, and F. Stephen Hodi, Dana-Farber Cancer Institute; Rebecca S. Heist and Lecia V. Sequist, Massachusetts General Hospital Cancer Center; David F. McDermott, Beth Israel Deaconess Medical Center, Boston, MA; Scott J. Antonia and Mary C. Pinder-Schenck, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Naiyer A. Rizvi, Richard D. Carvajal, and Matthew D. Hellmann, Memorial Sloan Kettering Cancer Center, New York, NY; John D. Powderly, Carolina BioOncology Institute, Huntersville, NC; David C. Smith, University of Michigan, Ann Arbor, MI; Philip Leming, Christ Hospital Cancer Center, Cincinnati, OH; Suzanne L. Topalian, Drew M. Pardoll, and Julie R. Brahmer, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, MD; and Vindira Sankar, Christoph M. Ahlers, Mark Salvati, Jon M. Wigginton, Georgia D. Kollia, and Ashok K. Gupta, Bristol-Myers Squibb, Princeton, NJ
| | - David F McDermott
- Scott N. Gettinger and Mario Sznol, Yale Cancer Center, New Haven, CT; Leora Horn, David P. Carbone, and Jeffrey A. Sosman, Vanderbilt University Medical Center; David R. Spigel, Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN; Leena Gandhi, David M. Jackman, and F. Stephen Hodi, Dana-Farber Cancer Institute; Rebecca S. Heist and Lecia V. Sequist, Massachusetts General Hospital Cancer Center; David F. McDermott, Beth Israel Deaconess Medical Center, Boston, MA; Scott J. Antonia and Mary C. Pinder-Schenck, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Naiyer A. Rizvi, Richard D. Carvajal, and Matthew D. Hellmann, Memorial Sloan Kettering Cancer Center, New York, NY; John D. Powderly, Carolina BioOncology Institute, Huntersville, NC; David C. Smith, University of Michigan, Ann Arbor, MI; Philip Leming, Christ Hospital Cancer Center, Cincinnati, OH; Suzanne L. Topalian, Drew M. Pardoll, and Julie R. Brahmer, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, MD; and Vindira Sankar, Christoph M. Ahlers, Mark Salvati, Jon M. Wigginton, Georgia D. Kollia, and Ashok K. Gupta, Bristol-Myers Squibb, Princeton, NJ
| | - Drew M Pardoll
- Scott N. Gettinger and Mario Sznol, Yale Cancer Center, New Haven, CT; Leora Horn, David P. Carbone, and Jeffrey A. Sosman, Vanderbilt University Medical Center; David R. Spigel, Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN; Leena Gandhi, David M. Jackman, and F. Stephen Hodi, Dana-Farber Cancer Institute; Rebecca S. Heist and Lecia V. Sequist, Massachusetts General Hospital Cancer Center; David F. McDermott, Beth Israel Deaconess Medical Center, Boston, MA; Scott J. Antonia and Mary C. Pinder-Schenck, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Naiyer A. Rizvi, Richard D. Carvajal, and Matthew D. Hellmann, Memorial Sloan Kettering Cancer Center, New York, NY; John D. Powderly, Carolina BioOncology Institute, Huntersville, NC; David C. Smith, University of Michigan, Ann Arbor, MI; Philip Leming, Christ Hospital Cancer Center, Cincinnati, OH; Suzanne L. Topalian, Drew M. Pardoll, and Julie R. Brahmer, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, MD; and Vindira Sankar, Christoph M. Ahlers, Mark Salvati, Jon M. Wigginton, Georgia D. Kollia, and Ashok K. Gupta, Bristol-Myers Squibb, Princeton, NJ
| | - Vindira Sankar
- Scott N. Gettinger and Mario Sznol, Yale Cancer Center, New Haven, CT; Leora Horn, David P. Carbone, and Jeffrey A. Sosman, Vanderbilt University Medical Center; David R. Spigel, Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN; Leena Gandhi, David M. Jackman, and F. Stephen Hodi, Dana-Farber Cancer Institute; Rebecca S. Heist and Lecia V. Sequist, Massachusetts General Hospital Cancer Center; David F. McDermott, Beth Israel Deaconess Medical Center, Boston, MA; Scott J. Antonia and Mary C. Pinder-Schenck, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Naiyer A. Rizvi, Richard D. Carvajal, and Matthew D. Hellmann, Memorial Sloan Kettering Cancer Center, New York, NY; John D. Powderly, Carolina BioOncology Institute, Huntersville, NC; David C. Smith, University of Michigan, Ann Arbor, MI; Philip Leming, Christ Hospital Cancer Center, Cincinnati, OH; Suzanne L. Topalian, Drew M. Pardoll, and Julie R. Brahmer, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, MD; and Vindira Sankar, Christoph M. Ahlers, Mark Salvati, Jon M. Wigginton, Georgia D. Kollia, and Ashok K. Gupta, Bristol-Myers Squibb, Princeton, NJ
| | - Christoph M Ahlers
- Scott N. Gettinger and Mario Sznol, Yale Cancer Center, New Haven, CT; Leora Horn, David P. Carbone, and Jeffrey A. Sosman, Vanderbilt University Medical Center; David R. Spigel, Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN; Leena Gandhi, David M. Jackman, and F. Stephen Hodi, Dana-Farber Cancer Institute; Rebecca S. Heist and Lecia V. Sequist, Massachusetts General Hospital Cancer Center; David F. McDermott, Beth Israel Deaconess Medical Center, Boston, MA; Scott J. Antonia and Mary C. Pinder-Schenck, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Naiyer A. Rizvi, Richard D. Carvajal, and Matthew D. Hellmann, Memorial Sloan Kettering Cancer Center, New York, NY; John D. Powderly, Carolina BioOncology Institute, Huntersville, NC; David C. Smith, University of Michigan, Ann Arbor, MI; Philip Leming, Christ Hospital Cancer Center, Cincinnati, OH; Suzanne L. Topalian, Drew M. Pardoll, and Julie R. Brahmer, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, MD; and Vindira Sankar, Christoph M. Ahlers, Mark Salvati, Jon M. Wigginton, Georgia D. Kollia, and Ashok K. Gupta, Bristol-Myers Squibb, Princeton, NJ
| | - Mark Salvati
- Scott N. Gettinger and Mario Sznol, Yale Cancer Center, New Haven, CT; Leora Horn, David P. Carbone, and Jeffrey A. Sosman, Vanderbilt University Medical Center; David R. Spigel, Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN; Leena Gandhi, David M. Jackman, and F. Stephen Hodi, Dana-Farber Cancer Institute; Rebecca S. Heist and Lecia V. Sequist, Massachusetts General Hospital Cancer Center; David F. McDermott, Beth Israel Deaconess Medical Center, Boston, MA; Scott J. Antonia and Mary C. Pinder-Schenck, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Naiyer A. Rizvi, Richard D. Carvajal, and Matthew D. Hellmann, Memorial Sloan Kettering Cancer Center, New York, NY; John D. Powderly, Carolina BioOncology Institute, Huntersville, NC; David C. Smith, University of Michigan, Ann Arbor, MI; Philip Leming, Christ Hospital Cancer Center, Cincinnati, OH; Suzanne L. Topalian, Drew M. Pardoll, and Julie R. Brahmer, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, MD; and Vindira Sankar, Christoph M. Ahlers, Mark Salvati, Jon M. Wigginton, Georgia D. Kollia, and Ashok K. Gupta, Bristol-Myers Squibb, Princeton, NJ
| | - Jon M Wigginton
- Scott N. Gettinger and Mario Sznol, Yale Cancer Center, New Haven, CT; Leora Horn, David P. Carbone, and Jeffrey A. Sosman, Vanderbilt University Medical Center; David R. Spigel, Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN; Leena Gandhi, David M. Jackman, and F. Stephen Hodi, Dana-Farber Cancer Institute; Rebecca S. Heist and Lecia V. Sequist, Massachusetts General Hospital Cancer Center; David F. McDermott, Beth Israel Deaconess Medical Center, Boston, MA; Scott J. Antonia and Mary C. Pinder-Schenck, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Naiyer A. Rizvi, Richard D. Carvajal, and Matthew D. Hellmann, Memorial Sloan Kettering Cancer Center, New York, NY; John D. Powderly, Carolina BioOncology Institute, Huntersville, NC; David C. Smith, University of Michigan, Ann Arbor, MI; Philip Leming, Christ Hospital Cancer Center, Cincinnati, OH; Suzanne L. Topalian, Drew M. Pardoll, and Julie R. Brahmer, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, MD; and Vindira Sankar, Christoph M. Ahlers, Mark Salvati, Jon M. Wigginton, Georgia D. Kollia, and Ashok K. Gupta, Bristol-Myers Squibb, Princeton, NJ
| | - Matthew D Hellmann
- Scott N. Gettinger and Mario Sznol, Yale Cancer Center, New Haven, CT; Leora Horn, David P. Carbone, and Jeffrey A. Sosman, Vanderbilt University Medical Center; David R. Spigel, Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN; Leena Gandhi, David M. Jackman, and F. Stephen Hodi, Dana-Farber Cancer Institute; Rebecca S. Heist and Lecia V. Sequist, Massachusetts General Hospital Cancer Center; David F. McDermott, Beth Israel Deaconess Medical Center, Boston, MA; Scott J. Antonia and Mary C. Pinder-Schenck, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Naiyer A. Rizvi, Richard D. Carvajal, and Matthew D. Hellmann, Memorial Sloan Kettering Cancer Center, New York, NY; John D. Powderly, Carolina BioOncology Institute, Huntersville, NC; David C. Smith, University of Michigan, Ann Arbor, MI; Philip Leming, Christ Hospital Cancer Center, Cincinnati, OH; Suzanne L. Topalian, Drew M. Pardoll, and Julie R. Brahmer, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, MD; and Vindira Sankar, Christoph M. Ahlers, Mark Salvati, Jon M. Wigginton, Georgia D. Kollia, and Ashok K. Gupta, Bristol-Myers Squibb, Princeton, NJ
| | - Georgia D Kollia
- Scott N. Gettinger and Mario Sznol, Yale Cancer Center, New Haven, CT; Leora Horn, David P. Carbone, and Jeffrey A. Sosman, Vanderbilt University Medical Center; David R. Spigel, Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN; Leena Gandhi, David M. Jackman, and F. Stephen Hodi, Dana-Farber Cancer Institute; Rebecca S. Heist and Lecia V. Sequist, Massachusetts General Hospital Cancer Center; David F. McDermott, Beth Israel Deaconess Medical Center, Boston, MA; Scott J. Antonia and Mary C. Pinder-Schenck, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Naiyer A. Rizvi, Richard D. Carvajal, and Matthew D. Hellmann, Memorial Sloan Kettering Cancer Center, New York, NY; John D. Powderly, Carolina BioOncology Institute, Huntersville, NC; David C. Smith, University of Michigan, Ann Arbor, MI; Philip Leming, Christ Hospital Cancer Center, Cincinnati, OH; Suzanne L. Topalian, Drew M. Pardoll, and Julie R. Brahmer, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, MD; and Vindira Sankar, Christoph M. Ahlers, Mark Salvati, Jon M. Wigginton, Georgia D. Kollia, and Ashok K. Gupta, Bristol-Myers Squibb, Princeton, NJ
| | - Ashok K Gupta
- Scott N. Gettinger and Mario Sznol, Yale Cancer Center, New Haven, CT; Leora Horn, David P. Carbone, and Jeffrey A. Sosman, Vanderbilt University Medical Center; David R. Spigel, Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN; Leena Gandhi, David M. Jackman, and F. Stephen Hodi, Dana-Farber Cancer Institute; Rebecca S. Heist and Lecia V. Sequist, Massachusetts General Hospital Cancer Center; David F. McDermott, Beth Israel Deaconess Medical Center, Boston, MA; Scott J. Antonia and Mary C. Pinder-Schenck, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Naiyer A. Rizvi, Richard D. Carvajal, and Matthew D. Hellmann, Memorial Sloan Kettering Cancer Center, New York, NY; John D. Powderly, Carolina BioOncology Institute, Huntersville, NC; David C. Smith, University of Michigan, Ann Arbor, MI; Philip Leming, Christ Hospital Cancer Center, Cincinnati, OH; Suzanne L. Topalian, Drew M. Pardoll, and Julie R. Brahmer, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, MD; and Vindira Sankar, Christoph M. Ahlers, Mark Salvati, Jon M. Wigginton, Georgia D. Kollia, and Ashok K. Gupta, Bristol-Myers Squibb, Princeton, NJ
| | - Julie R Brahmer
- Scott N. Gettinger and Mario Sznol, Yale Cancer Center, New Haven, CT; Leora Horn, David P. Carbone, and Jeffrey A. Sosman, Vanderbilt University Medical Center; David R. Spigel, Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN; Leena Gandhi, David M. Jackman, and F. Stephen Hodi, Dana-Farber Cancer Institute; Rebecca S. Heist and Lecia V. Sequist, Massachusetts General Hospital Cancer Center; David F. McDermott, Beth Israel Deaconess Medical Center, Boston, MA; Scott J. Antonia and Mary C. Pinder-Schenck, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL; Naiyer A. Rizvi, Richard D. Carvajal, and Matthew D. Hellmann, Memorial Sloan Kettering Cancer Center, New York, NY; John D. Powderly, Carolina BioOncology Institute, Huntersville, NC; David C. Smith, University of Michigan, Ann Arbor, MI; Philip Leming, Christ Hospital Cancer Center, Cincinnati, OH; Suzanne L. Topalian, Drew M. Pardoll, and Julie R. Brahmer, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, MD; and Vindira Sankar, Christoph M. Ahlers, Mark Salvati, Jon M. Wigginton, Georgia D. Kollia, and Ashok K. Gupta, Bristol-Myers Squibb, Princeton, NJ
| |
Collapse
|
4211
|
Abstract
The toxicities of immunotherapy for cancer are as diverse as the type of treatments that have been devised. These range from cytokine therapies that induce capillary leakage to vaccines associated with low levels of autoimmunity to cell therapies that can induce damaging cross-reactivity with normal tissue to checkpoint protein inhibitors that induce immune-related adverse events that are autoinflammatory in nature. The thread that ties these toxicities together is their mechanism-based immune nature and the T-cell-mediated adverse events seen. The basis for the majority of these adverse events is a hyperactivated T-cell response with reactivity directed against normal tissue, resulting in the generation of high levels of CD4 T-helper cell cytokines or increased migration of cytolytic CD8 T cells within normal tissues. The T-cell immune response is not tissue specific and may reflect a diffuse expansion of the T-cell repertoire that induces cross-reactivity with normal tissue, effectively breaking tolerance that is active with cytokines, vaccines, and checkpoint protein inhibitors and passive in the case of adoptive cell therapy. Cytokines seem to generate diffuse and nonspecific T-cell reactivity, whereas checkpoint protein inhibition, vaccines, and adoptive cell therapy seem to activate more specific T cells that interact directly with normal tissues, potentially causing specific organ damage. In this review, we summarize the toxicities that are unique to immunotherapies, emphasizing the need to familiarize the oncology practitioner with the spectrum of adverse events seen with newly approved and emerging modalities.
Collapse
Affiliation(s)
- Jeffrey S Weber
- Jeffrey S. Weber, Moffitt Cancer Center, Tampa, FL; James C. Yang, National Cancer Institute, Bethesda, MD; Michael B. Atkins, Lombardi Cancer Center, Georgetown University, Washington, DC; and Mary L. Disis, The Fred Hutchinson Cancer Center, University of Washington, Seattle, WA.
| | - James C Yang
- Jeffrey S. Weber, Moffitt Cancer Center, Tampa, FL; James C. Yang, National Cancer Institute, Bethesda, MD; Michael B. Atkins, Lombardi Cancer Center, Georgetown University, Washington, DC; and Mary L. Disis, The Fred Hutchinson Cancer Center, University of Washington, Seattle, WA
| | - Michael B Atkins
- Jeffrey S. Weber, Moffitt Cancer Center, Tampa, FL; James C. Yang, National Cancer Institute, Bethesda, MD; Michael B. Atkins, Lombardi Cancer Center, Georgetown University, Washington, DC; and Mary L. Disis, The Fred Hutchinson Cancer Center, University of Washington, Seattle, WA
| | - Mary L Disis
- Jeffrey S. Weber, Moffitt Cancer Center, Tampa, FL; James C. Yang, National Cancer Institute, Bethesda, MD; Michael B. Atkins, Lombardi Cancer Center, Georgetown University, Washington, DC; and Mary L. Disis, The Fred Hutchinson Cancer Center, University of Washington, Seattle, WA
| |
Collapse
|
4212
|
Carbognin L, Pilotto S, Milella M, Vaccaro V, Brunelli M, Caliò A, Cuppone F, Sperduti I, Giannarelli D, Chilosi M, Bronte V, Scarpa A, Bria E, Tortora G. Differential Activity of Nivolumab, Pembrolizumab and MPDL3280A according to the Tumor Expression of Programmed Death-Ligand-1 (PD-L1): Sensitivity Analysis of Trials in Melanoma, Lung and Genitourinary Cancers. PLoS One 2015; 10:e0130142. [PMID: 26086854 PMCID: PMC4472786 DOI: 10.1371/journal.pone.0130142] [Citation(s) in RCA: 376] [Impact Index Per Article: 37.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Accepted: 05/18/2015] [Indexed: 12/13/2022] Open
Abstract
Background The potential predictive role of programmed death-ligand-1 (PD-L1) expression on tumor cells in the context of solid tumor treated with checkpoint inhibitors targeting the PD-1 pathway represents an issue for clinical research. Methods Overall response rate (ORR) was extracted from phase I-III trials investigating nivolumab, pembrolizumab and MPDL3280A for advanced melanoma, non-small cell lung cancer (NSCLC) and genitourinary cancer, and cumulated by adopting a fixed and random-effect model with 95% confidence interval (CI). Interaction test according to tumor PD-L1 was accomplished. A sensitivity analysis according to adopted drug, tumor type, PD-L1 cut-off and treatment line was performed. Results Twenty trials (1,475 patients) were identified. A significant interaction (p<0.0001) according to tumor PD-L1 expression was found in the overall sample with an ORR of 34.1% (95% CI 27.6-41.3%) in the PD-L1 positive and 19.9% (95% CI 15.4-25.3%) in the PD-L1 negative population. ORR was significantly higher in PD-L1 positive in comparison to PD-L1 negative patients for nivolumab and pembrolizumab, with an absolute difference of 16.4% and 19.5%, respectively. A significant difference in activity of 22.8% and 8.7% according to PD-L1 was found for melanoma and NSCLC, respectively, with no significant difference for genitourinary cancer. Conclusion Overall, the three antibodies provide a significant differential effect in terms of activity according to PD-L1 expression on tumor cells. The predictive value of PD-L1 on tumor cells seems to be more robust for anti-PD-1 antibody (nivolumab and pembrolizumab), and in the context of advanced melanoma and NSCLC.
Collapse
Affiliation(s)
- Luisa Carbognin
- Medical Oncology, University of Verona, Azienda Ospedaliera Universitaria Integrata, Verona, Italy
| | - Sara Pilotto
- Medical Oncology, University of Verona, Azienda Ospedaliera Universitaria Integrata, Verona, Italy
| | - Michele Milella
- Medical Oncology, Regina Elena National Cancer Institute, Roma, Italy
| | - Vanja Vaccaro
- Medical Oncology, Regina Elena National Cancer Institute, Roma, Italy
| | - Matteo Brunelli
- Department of Pathology and Diagnostic, University of Verona, Azienda Ospedaliera Universitaria Integrata, Verona, Italy
| | - Anna Caliò
- Department of Pathology and Diagnostic, University of Verona, Azienda Ospedaliera Universitaria Integrata, Verona, Italy
| | | | | | | | - Marco Chilosi
- Department of Pathology and Diagnostic, University of Verona, Azienda Ospedaliera Universitaria Integrata, Verona, Italy
| | - Vincenzo Bronte
- Department of Pathology and Diagnostic, University of Verona, Azienda Ospedaliera Universitaria Integrata, Verona, Italy
| | - Aldo Scarpa
- Department of Pathology and Diagnostic, University of Verona, Azienda Ospedaliera Universitaria Integrata, Verona, Italy
- ARC-NET Center for Applied Research on Cancer, Verona, Italy
| | - Emilio Bria
- Medical Oncology, University of Verona, Azienda Ospedaliera Universitaria Integrata, Verona, Italy
- * E-mail:
| | - Giampaolo Tortora
- Medical Oncology, University of Verona, Azienda Ospedaliera Universitaria Integrata, Verona, Italy
| |
Collapse
|
4213
|
Lollini PL, Cavallo F, Nanni P, Quaglino E. The Promise of Preventive Cancer Vaccines. Vaccines (Basel) 2015; 3:467-89. [PMID: 26343198 PMCID: PMC4494347 DOI: 10.3390/vaccines3020467] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 06/04/2015] [Accepted: 06/08/2015] [Indexed: 01/01/2023] Open
Abstract
Years of unsuccessful attempts at fighting established tumors with vaccines have taught us all that they are only able to truly impact patient survival when used in a preventive setting, as would normally be the case for traditional vaccines against infectious diseases. While true primary cancer prevention is still but a long-term goal, secondary and tertiary prevention are already in the clinic and providing encouraging results. A combination of immunopreventive cancer strategies and recently approved checkpoint inhibitors is a further promise of forthcoming successful cancer disease control, but prevention will require a considerable reduction of currently reported toxicities. These considerations summed with the increased understanding of tumor antigens allow space for an optimistic view of the future.
Collapse
Affiliation(s)
- Pier-Luigi Lollini
- Department of Experimental Diagnostic and Specialty Medicine (DIMES), University of Bologna, Viale Filopanti 22, Bologna 40126, Italy.
| | - Federica Cavallo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Via Nizza 52, Torino 10126, Italy.
| | - Patrizia Nanni
- Department of Experimental Diagnostic and Specialty Medicine (DIMES), University of Bologna, Viale Filopanti 22, Bologna 40126, Italy.
| | - Elena Quaglino
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Via Nizza 52, Torino 10126, Italy.
| |
Collapse
|
4214
|
Ito A, Kondo S, Tada K, Kitano S. Clinical Development of Immune Checkpoint Inhibitors. BIOMED RESEARCH INTERNATIONAL 2015; 2015:605478. [PMID: 26161407 PMCID: PMC4486755 DOI: 10.1155/2015/605478] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Accepted: 12/07/2014] [Indexed: 01/21/2023]
Abstract
Recent progress in cancer immunotherapy has been remarkable. Most striking are the clinical development and approval of immunomodulators, also known as immune checkpoint inhibitors. These monoclonal antibodies (mAb) are directed to immune checkpoint molecules, which are expressed on immune cells and mediate signals to attenuate excessive immune reactions. Although mAbs targeting tumor associated antigens, such as anti-CD20 mAb and anti-Her2 mAb, directly recognize tumor cells and induce cell death, immune checkpoint inhibitors restore and augment the antitumor immune activities of cytotoxic T cells by blocking immune checkpoint molecules on T cells or their ligands on antigen presenting and tumor cells. Based on preclinical data, many clinical trials have demonstrated the acceptable safety profiles and efficacies of immune checkpoint inhibitors in a variety of cancers. The first in class approved immune checkpoint inhibitor is ipilimumab, an anti-CTLA-4 (cytotoxic T lymphocyte antigen-4) mAb. Two pivotal phase III randomized controlled trials demonstrated a survival benefit in patients with metastatic melanoma. In 2011, the US Food and Drug Administration (FDA) approved ipilimumab for metastatic melanoma. Several clinical trials have since investigated new agents, alone and in combination, for various cancers. In this review, we discuss the current development status of and future challenges in utilizing immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Ayumu Ito
- Department of Hematopoietic Stem Cell Transplantation, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
| | - Shunsuke Kondo
- Department of Experimental Therapeutics, Exploratory Oncology Research and Clinical Trial Center (EPOC), National Cancer Center, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
| | - Kohei Tada
- Division of Cancer Immunotherapy, Exploratory Oncology Research and Clinical Trial Center (EPOC), National Cancer Center, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
| | - Shigehisa Kitano
- Department of Experimental Therapeutics, Exploratory Oncology Research and Clinical Trial Center (EPOC), National Cancer Center, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
- Division of Cancer Immunotherapy, Exploratory Oncology Research and Clinical Trial Center (EPOC), National Cancer Center, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
| |
Collapse
|
4215
|
Page DB, Bourla AB, Daniyan A, Naidoo J, Smith E, Smith M, Friedman C, Khalil DN, Funt S, Shoushtari AN, Overwijk WW, Sharma P, Callahan MK. Tumor immunology and cancer immunotherapy: summary of the 2014 SITC primer. J Immunother Cancer 2015. [PMCID: PMC4469248 DOI: 10.1186/s40425-015-0072-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
|
4216
|
Pico de Coaña Y, Choudhury A, Kiessling R. Checkpoint blockade for cancer therapy: revitalizing a suppressed immune system. Trends Mol Med 2015; 21:482-91. [PMID: 26091825 DOI: 10.1016/j.molmed.2015.05.005] [Citation(s) in RCA: 134] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Revised: 05/19/2015] [Accepted: 05/20/2015] [Indexed: 01/22/2023]
Abstract
Immune checkpoint receptors are crucial molecules for fine-tuning immune responses. Checkpoint signaling dampens T cell activation to avoid autoimmunity and the destructive effects of an excessive inflammatory response. It is well established that tumors use several mechanisms to avoid elimination by the immune system, and one involves hijacking these checkpoint pathways. Checkpoint blockade therapy utilizes monoclonal antibodies to release the brakes from suppressed T cells, allowing them to be activated and recover their antitumor activity. This therapeutic approach has revolutionized cancer immunotherapy, and extraordinary increases in overall survival were noted, first with anti-CTLA-4 (cytotoxic T lymphocyte-associated protein 4) and subsequently with anti-PD-1 (programmed cell death receptor-1) in melanoma and other malignancies.
Collapse
Affiliation(s)
- Yago Pico de Coaña
- Department of Oncology and Pathology, Cancer Center Karolinska, Karolinska Institutet, Stockholm, Sweden.
| | | | - Rolf Kiessling
- Department of Oncology and Pathology, Cancer Center Karolinska, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
4217
|
Abstract
Scientific advances in the last decade have demonstrated the critical role of host immune system in the elimination and suppression of cancer cells. Better knowledge of signaling pathways has enabled the development of new cancer immunotherapy. The discovery of negative feedback mechanisms following the lymphocyte activation has promoted the development of new antibodies targeting molecule inhibitors such as PD1, PDL1 or CTLA-4. Dramatic results were obtained with melanoma. Checkpoint inhibitors (pembrolizumab and ipilimumab) have many advantages in terms of rate of objective response and overall survival. Recent studies in translational research aimed to understand and analyze mechanisms of action of anti-PD1/anti-PDL1. Expression of PDL1 in the tumor is associated with a significantly greater objective response rate (immunohistochemistry). Nevertheless, limits with tumor immunohistochemical analysis encourage new biomarkers research. Other immunotherapy approaches, such as cell and gene therapies using engineered T cells call for further advancements to broaden their applicability. However, these therapies are very expensive and their manufacturing process very restrictive, which could lately limit their use in case of inefficiency of checkpoint inhibitors or when lymphocytic infiltration in tumor is absent. In this case, the objective would be to engineer ex vivo the patient's immune system by restoring the ability of T cells to identify and suppress tumor cells. Currently, two gene-reprogramming tools are under development: chimeric antigen receptor and TCR modified T cells.
Collapse
|
4218
|
Pennock GK, Chow LQM. The Evolving Role of Immune Checkpoint Inhibitors in Cancer Treatment. Oncologist 2015; 20:812-22. [PMID: 26069281 DOI: 10.1634/theoncologist.2014-0422] [Citation(s) in RCA: 184] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2014] [Accepted: 03/20/2015] [Indexed: 01/12/2023] Open
Abstract
UNLABELLED Traditional treatment modalities for advanced cancer (radiotherapy, chemotherapy, or targeted agents) act directly on tumors to inhibit or destroy them. Along with surgery, these modalities are predominantly palliative, with toxicity and only modest improvements in survival in patients with advanced solid tumors. Accordingly, long-term survival rates for most patients with advanced cancer remain low, thus there is a need for cancer treatments with favorable benefit and toxicity profiles that can potentially result in long-term survival. The immune system plays a critical role in the recognition and eradication of tumor cells ("immune surveillance"), and immunotherapies based on this concept have been used for decades with some success against a few tumor types; however, most immunotherapies were limited by a lack of either substantial efficacy or specificity, resulting in toxicity. We now have a greater understanding of the complex interactions between the immune system and tumors and have identified key molecules that govern these interactions. This information has revitalized the interest in immunotherapy as an evolving treatment modality using immunotherapeutics designed to overcome the mechanisms exploited by tumors to evade immune destruction. Immunotherapies have potentially complementary mechanisms of action that may allow them to be combined with other immunotherapeutics, chemotherapy, targeted therapy, or other traditional therapies. This review discusses the concepts and data behind immunotherapies, with a focus on the checkpoint inhibitors and their responses, toxicities, and potential for long-term survival, and explores promising single-agent and combination therapies in development. IMPLICATIONS FOR PRACTICE Immunotherapy is an evolving treatment approach based on the role of the immune system in eradicating cancer. An example of an immunotherapeutic is ipilimumab, an antibody that blocks cytotoxic T-lymphocyte antigen-4 (CTLA-4) to augment antitumor immune responses. Ipilimumab is approved for advanced melanoma and induced long-term survival in a proportion of patients. The programmed death-1 (PD-1) checkpoint inhibitors are promising immunotherapies with demonstrated sustained antitumor responses in several tumors. Because they harness the patient's own immune system, immunotherapies have the potential to be a powerful weapon against cancer.
Collapse
Affiliation(s)
- Gregory K Pennock
- Levine Cancer Institute, Charlotte, North Carolina, USA; University of Washington, Seattle, Washington, USA
| | - Laura Q M Chow
- Levine Cancer Institute, Charlotte, North Carolina, USA; University of Washington, Seattle, Washington, USA
| |
Collapse
|
4219
|
LoRusso PM, Boerner SA, Pilat MJ, Forman KM, Zuccaro CY, Kiefer JA, Liang WS, Hunsberger S, Redman BG, Markovic SN, Sekulic A, Bryce AH, Joseph RW, Cowey CL, Fecher LA, Sosman JA, Chapman PB, Schwartz GK, Craig DW, Carpten JD, Trent JM. Pilot Trial of Selecting Molecularly Guided Therapy for Patients with Non-V600 BRAF-Mutant Metastatic Melanoma: Experience of the SU2C/MRA Melanoma Dream Team. Mol Cancer Ther 2015; 14:1962-71. [PMID: 26063764 DOI: 10.1158/1535-7163.mct-15-0153] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 05/21/2015] [Indexed: 01/06/2023]
Abstract
Targeted therapies and immunotherapies have led to significant improvements in the treatment of advanced cancers, including metastatic melanoma. However, new strategies are desperately needed to overcome therapeutic resistance to these agents, as well as to identify effective treatment approaches for cancer patients that fall outside major targetable mutational subtypes (e.g., non-V600 BRAF melanoma). One such strategy is to extend the paradigm of individually tailored, molecularly targeted therapy into a broader spectrum of melanoma patients, particularly those bearing tumors without commonly recognized therapeutic targets, as well as having failed or were ineligible for immunotherapy. In this nontreatment pilot study, next-generation sequencing (NGS) technologies were utilized, including whole genome and whole transcriptome sequencing, to identify molecular aberrations in patients with non-V600 BRAF metastatic melanoma. This information was then rationally matched to an appropriate clinical treatment from a defined pharmacopeia. Five patients with advanced non-V600 BRAF metastatic melanoma were enrolled. We demonstrated successful performance of the following during a clinically relevant time period: patient tumor biopsy, quality DNA/RNA extraction, DNA/RNA-based sequencing for gene expression analysis, analysis utilizing a series of data integration methodologies, report generation, and tumor board review with formulated treatment plan. Streamlining measures were conducted based on the experiences of enrolling, collecting specimens, and analyzing the molecular signatures of patients. We demonstrated the feasibility of using NGS to identify molecular aberrations and generate an individualized treatment plan in this patient population. A randomized treatment study utilizing lessons learned from the conduct of this pilot study is currently underway.
Collapse
Affiliation(s)
- Patricia M LoRusso
- Yale Cancer Center, New Haven, Connecticut. Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, Michigan.
| | - Scott A Boerner
- Yale Cancer Center, New Haven, Connecticut. Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, Michigan
| | - Mary Jo Pilat
- Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, Michigan
| | - Karen M Forman
- Yale Cancer Center, New Haven, Connecticut. Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, Michigan
| | - Clarice Y Zuccaro
- Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, Michigan
| | | | - Winnie S Liang
- Translational Genomics Research Institute, Phoenix, Arizona
| | - Sally Hunsberger
- Biometrics Research Branch, National Cancer Institute, National Institutes of Health, Rockville, Maryland
| | - Bruce G Redman
- University of Michigan Comprehensive Cancer Center (UMCCC), Ann Arbor, Michigan
| | | | | | | | | | - C Lance Cowey
- Charles A. Sammons Cancer Center/Baylor University Medical Center, Dallas, Texas
| | - Leslie Anne Fecher
- University of Michigan Comprehensive Cancer Center (UMCCC), Ann Arbor, Michigan
| | | | - Paul B Chapman
- Memorial Sloan-Kettering Cancer Center, New York, New York
| | | | - David W Craig
- Translational Genomics Research Institute, Phoenix, Arizona
| | - John D Carpten
- Translational Genomics Research Institute, Phoenix, Arizona
| | | |
Collapse
|
4220
|
Lipson EJ, Forde PM, Hammers HJ, Emens LA, Taube JM, Topalian SL. Antagonists of PD-1 and PD-L1 in Cancer Treatment. Semin Oncol 2015; 42:587-600. [PMID: 26320063 DOI: 10.1053/j.seminoncol.2015.05.013] [Citation(s) in RCA: 235] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The PD-1 pathway, comprising the immune cell co-receptor Programmed Death 1 (PD-1) and its ligands, PD-L1 (B7-H1) and PD-L2 (B7-DC), mediates local immunosuppression in the tumor microenvironment. Drugs designed to block PD-1 or PD-L1 "release the brakes" on anti-tumor immunity and have demonstrated clinical activity in several types of advanced cancers, validating this pathway as a target for cancer therapy. Two such drugs have recently been approved to treat melanoma and lung cancers, and regulatory approvals in first- and second-line settings for additional cancer types are anticipated. The manageable safety profile of PD-1/PD-L1 blocking drugs identifies them as suitable for outpatient administration and the development of combinatorial therapies. Ongoing studies aim to identify biomarkers to guide patient selection, which would further improve the risk:benefit ratio for these drugs.
Collapse
Affiliation(s)
- Evan J Lipson
- Department of Oncology, The Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | - Patrick M Forde
- Department of Oncology, The Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | - Hans-Joerg Hammers
- Department of Oncology, The Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | - Leisha A Emens
- Department of Oncology, The Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | - Janis M Taube
- Department of Oncology, The Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD; Department of Dermatology, The Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD; Department of Pathology, The Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | - Suzanne L Topalian
- Department of Surgery, The Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| |
Collapse
|
4221
|
Affiliation(s)
- Antoni Ribas
- University of California, Los Angeles, and the Jonsson Comprehensive Cancer Center, Los Angeles, CA
| | | |
Collapse
|
4222
|
Novel Approaches to Treatment of Advanced Melanoma: A Review on Targeted Therapy and Immunotherapy. BIOMED RESEARCH INTERNATIONAL 2015; 2015:851387. [PMID: 26171394 PMCID: PMC4478296 DOI: 10.1155/2015/851387] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2015] [Accepted: 05/19/2015] [Indexed: 01/22/2023]
Abstract
The incidence of malignant melanoma is increasing. The majority of patients are diagnosed in early stages when the disease is highly curable. However, the more advanced or metastatic cases have always been a challenge for clinicians. The poor prognosis for patients with melanoma is now changing as numerous of promising approaches have appeared recently. The discovery of aberrations of pathways responsible for intracellular signal transduction allowed us to introduce agents specifically targeting the mutated cascades. Numerous clinical studies have been conducted to improve effectiveness of melanoma treatment. From 2011 until now, the U.S. FDA has approved seven novel agents, such as BRAF-inhibitors (vemurafenib 2011, dabrafenib 2013), MEK-inhibitors (trametinib 2013), anti-PD1 antibodies (nivolumab 2014, pembrolizumab 2014), anti-CTLA-4 antibody (ipilimumab 2011), or peginterferon-alfa-2b (2011) intended to be used in most advanced cases of melanoma. Nevertheless, clinicians continue working on new possible methods of treatment as resistance to the novel drugs is a commonly observed problem. This paper is based on latest data published until the end of January 2015.
Collapse
|
4223
|
Lee JJ, Sholl LM, Lindeman NI, Granter SR, Laga AC, Shivdasani P, Chin G, Luke JJ, Ott PA, Hodi FS, Mihm MC, Lin JY, Werchniak AE, Haynes HA, Bailey N, Liu R, Murphy GF, Lian CG. Targeted next-generation sequencing reveals high frequency of mutations in epigenetic regulators across treatment-naïve patient melanomas. Clin Epigenetics 2015; 7:59. [PMID: 26221190 PMCID: PMC4517542 DOI: 10.1186/s13148-015-0091-3] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Accepted: 05/27/2015] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Recent developments in genomic sequencing have advanced our understanding of the mutations underlying human malignancy. Melanoma is a prototype of an aggressive, genetically heterogeneous cancer notorious for its biologic plasticity and predilection towards developing resistance to targeted therapies. Evidence is rapidly accumulating that dysregulated epigenetic mechanisms (DNA methylation/demethylation, histone modification, non-coding RNAs) may play a central role in the pathogenesis of melanoma. Therefore, we sought to characterize the frequency and nature of mutations in epigenetic regulators in clinical, treatment-naïve, patient melanoma specimens obtained from one academic institution. RESULTS Targeted next-generation sequencing for 275 known and investigative cancer genes (of which 41 genes, or 14.9 %, encoded an epigenetic regulator) of 38 treatment-naïve patient melanoma samples revealed that 22.3 % (165 of 740) of all non-silent mutations affected an epigenetic regulator. The most frequently mutated genes were BRAF, MECOM, NRAS, TP53, MLL2, and CDKN2A. Of the 40 most commonly mutated genes, 12 (30.0 %) encoded epigenetic regulators, including genes encoding enzymes involved in histone modification (MECOM, MLL2, SETD2), chromatin remodeling (ARID1B, ARID2), and DNA methylation and demethylation (TET2, IDH1). Among the 38 patient melanoma samples, 35 (92.1 %) harbored at least one mutation in an epigenetic regulator. The genes with the highest number of total UVB-signature mutations encoded epigenetic regulators, including MLL2 (100 %, 16 of 16) and MECOM (82.6 %, 19 of 23). Moreover, on average, epigenetic genes harbored a significantly greater number of UVB-signature mutations per gene than non-epigenetic genes (3.7 versus 2.4, respectively; p = 0.01). Bioinformatics analysis of The Cancer Genome Atlas (TCGA) melanoma mutation dataset also revealed a frequency of mutations in the 41 epigenetic genes comparable to that found within our cohort of patient melanoma samples. CONCLUSIONS Our study identified a high prevalence of somatic mutations in genes encoding epigenetic regulators, including those involved in DNA demethylation, histone modification, chromatin remodeling, and microRNA processing. Moreover, UVB-signature mutations were found more commonly among epigenetic genes than in non-epigenetic genes. Taken together, these findings further implicate epigenetic mechanisms, particularly those involving the chromatin-remodeling enzyme MECOM/EVI1 and histone-modifying enzyme MLL2, in the pathobiology of melanoma.
Collapse
Affiliation(s)
- Jonathan J. Lee
- />Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, 221 Longwood Avenue, EBRC Suite 401, Boston, MA 02115 USA
| | - Lynette M. Sholl
- />Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, 221 Longwood Avenue, EBRC Suite 401, Boston, MA 02115 USA
| | - Neal I. Lindeman
- />Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, 221 Longwood Avenue, EBRC Suite 401, Boston, MA 02115 USA
| | - Scott R. Granter
- />Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, 221 Longwood Avenue, EBRC Suite 401, Boston, MA 02115 USA
| | - Alvaro C. Laga
- />Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, 221 Longwood Avenue, EBRC Suite 401, Boston, MA 02115 USA
| | - Priyanka Shivdasani
- />Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, 221 Longwood Avenue, EBRC Suite 401, Boston, MA 02115 USA
| | - Gary Chin
- />Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, 221 Longwood Avenue, EBRC Suite 401, Boston, MA 02115 USA
| | - Jason J. Luke
- />Melanoma Center, Dana–Farber Cancer Institute, Harvard Medical School, 450 Brookline Ave., Boston, MA 02215-5450 USA
| | - Patrick A. Ott
- />Melanoma Center, Dana–Farber Cancer Institute, Harvard Medical School, 450 Brookline Ave., Boston, MA 02215-5450 USA
| | - F. Stephen Hodi
- />Melanoma Center, Dana–Farber Cancer Institute, Harvard Medical School, 450 Brookline Ave., Boston, MA 02215-5450 USA
| | - Martin C. Mihm
- />Melanoma Center, Dana–Farber Cancer Institute, Harvard Medical School, 450 Brookline Ave., Boston, MA 02215-5450 USA
| | - Jennifer Y. Lin
- />Melanoma Center, Dana–Farber Cancer Institute, Harvard Medical School, 450 Brookline Ave., Boston, MA 02215-5450 USA
| | - Andrew E. Werchniak
- />Melanoma Center, Dana–Farber Cancer Institute, Harvard Medical School, 450 Brookline Ave., Boston, MA 02215-5450 USA
| | - Harley A. Haynes
- />Melanoma Center, Dana–Farber Cancer Institute, Harvard Medical School, 450 Brookline Ave., Boston, MA 02215-5450 USA
| | - Nancy Bailey
- />Melanoma Center, Dana–Farber Cancer Institute, Harvard Medical School, 450 Brookline Ave., Boston, MA 02215-5450 USA
| | - Robert Liu
- />Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, 221 Longwood Avenue, EBRC Suite 401, Boston, MA 02115 USA
| | - George F. Murphy
- />Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, 221 Longwood Avenue, EBRC Suite 401, Boston, MA 02115 USA
| | - Christine G. Lian
- />Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, 221 Longwood Avenue, EBRC Suite 401, Boston, MA 02115 USA
| |
Collapse
|
4224
|
Abstract
Mortality owing to liver cancer has increased in the past 20 years, and the latest estimates indicate that the global health burden of this disease will continue to grow. Most patients with hepatocellular carcinoma (HCC) are still diagnosed at intermediate or advanced disease stages, where curative approaches are often not feasible. Among the treatment options available, the molecular targeted agent sorafenib is able to significantly increase overall survival in these patients. Thereafter, up to seven large, randomized phase III clinical trials investigating other molecular therapies in the first-line and second-line settings have failed to improve on the results observed with this agent. Potential reasons for this include intertumour heterogeneity, issues with trial design and a lack of predictive biomarkers of response. During the past 5 years, substantial advances in our knowledge of the human genome have provided a comprehensive picture of commonly mutated genes in patients with HCC. This knowledge has not yet influenced clinical decision-making or current clinical practice guidelines. In this Review the authors summarize the molecular concepts of progression, discuss the potential reasons for clinical trial failure and propose new concepts of drug development, which might lead to clinical implementation of emerging targeted agents.
Collapse
|
4225
|
Imai Y, Ayithan N, Wu X, Yuan Y, Wang L, Hwang ST. Cutting Edge: PD-1 Regulates Imiquimod-Induced Psoriasiform Dermatitis through Inhibition of IL-17A Expression by Innate γδ-Low T Cells. THE JOURNAL OF IMMUNOLOGY 2015; 195:421-5. [PMID: 26048148 DOI: 10.4049/jimmunol.1500448] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 05/14/2015] [Indexed: 02/06/2023]
Abstract
Programmed cell death 1 (PD-1) is a key regulatory molecule that has been targeted in human cancers, including melanoma. In clinical testing, Abs against PD-1 have resulted in psoriasiform dermatitis (PsD). To determine whether PD-1 regulates PsD, we compared skin responses of PD-1-deficient (PD-1KO) mice and wild-type (WT) controls in an imiquimod (IMQ)-induced murine model of psoriasis. PD-1KO mice showed severe epidermal hyperplasia, greater neutrophilic infiltration, and higher expression of Th17 cytokines (versus WT mice). IMQ exposure increased PD-1 expression by skin γδ-low (GDL) T cells and enhanced expression of PD-L1 by keratinocytes. Three-fold increases in the percentage of IL-17A(+) GDL T cells were observed in skin cell suspensions derived from IMQ-treated PD-1KO mice (versus WT controls), suggesting that the lack of PD-1 has a functional effect not only on αβ T cells, but also on GDL T cells, and that PD-1 may play a regulatory role in PsD.
Collapse
Affiliation(s)
- Yasutomo Imai
- Department of Dermatology, Medical College of Wisconsin, Milwaukee, WI 53226; and
| | - Natarajan Ayithan
- Department of Microbiology and Molecular Genetics, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Xuesong Wu
- Department of Dermatology, Medical College of Wisconsin, Milwaukee, WI 53226; and
| | - Ying Yuan
- Department of Microbiology and Molecular Genetics, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Li Wang
- Department of Microbiology and Molecular Genetics, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Sam T Hwang
- Department of Dermatology, Medical College of Wisconsin, Milwaukee, WI 53226; and
| |
Collapse
|
4226
|
Kissick HT, Sanda MG. The role of active vaccination in cancer immunotherapy: lessons from clinical trials. Curr Opin Immunol 2015; 35:15-22. [PMID: 26050634 DOI: 10.1016/j.coi.2015.05.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Revised: 05/06/2015] [Accepted: 05/08/2015] [Indexed: 01/05/2023]
Abstract
In the past few years, a number of different immunotherapeutic strategies have shown impressive results in cancer patients. These successful approaches include blockade of immunosuppressive molecules like PD-1 and CTLA-4, adoptive transfer of patient derived and genetically modified T-cells, and vaccines that stimulate tumor antigen specific T-cells. However, several large vaccine trials recently failed to reach designated primary endpoints. In light of the success of other immunotherapeutic approaches, these negative results raise the questions of why vaccines have not generated a better response, and what the role of active vaccination will be moving forward in cancer immunotherapy.
Collapse
Affiliation(s)
- Haydn T Kissick
- Department of Urology, Emory University School of Medicine, United States; Department of Microbiology and Immunology, Emory University School of Medicine, United States.
| | - Martin G Sanda
- Department of Urology, Emory University School of Medicine, United States
| |
Collapse
|
4227
|
Page DB, Naidoo J, McArthur HL. The 2014 San Antonio Breast Cancer Symposium: A successful lift-off for breast immunotherapy? NPJ Breast Cancer 2015. [PMCID: PMC5515199 DOI: 10.1038/npjbcancer.2015.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Therapeutic immune checkpoint antibodies promote potentially durable cancer control by modulating key regulatory factors of the endogenous anti-tumor immune response. The first clinical trial data of these agents in breast cancer were presented at the 2014 San Antonio Breast Cancer Symposium, with seemingly modest response rates compared with metastatic melanoma and Hodgkin’s lymphoma. In this article, we review the San Antonio immunotherapy data, drawing key analogies to historical experiences in metastatic melanoma that support an enthusiastic outlook for immunotherapy in breast cancer.
Collapse
|
4228
|
Sunshine J, Taube JM. PD-1/PD-L1 inhibitors. Curr Opin Pharmacol 2015; 23:32-8. [PMID: 26047524 DOI: 10.1016/j.coph.2015.05.011] [Citation(s) in RCA: 441] [Impact Index Per Article: 44.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Revised: 05/06/2015] [Accepted: 05/15/2015] [Indexed: 12/26/2022]
Abstract
Tumors may adopt normal physiologic checkpoints for immunomodulation leading to an imbalance between tumor growth and host surveillance. Antibodies targeting the PD-1/PD-L1 checkpoint have shown dynamic and durable tumor regressions, suggesting a rebalancing of the host-tumor interaction. Nivolumab and pembrolizumab are the anti-PD-1 antibodies that are currently the furthest in clinical development, and anti-PD-L1 agents under investigation include MPDL3280A, MEDI4736, and BMS-936559. These agents have been used to treat advanced melanoma, non-small cell lung cancer, renal cell carcinoma, bladder cancer and Hodgkin lymphoma, amongst other tumor types. In this article, we review the updated response results for early clinical trials, note recent FDA actions regarding this class of agents, and summarize results across trials looking at PD-L1 status as a predictor of response to anti-PD-1/PD-L1.
Collapse
Affiliation(s)
- Joel Sunshine
- Department of Dermatology, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Janis M Taube
- Department of Dermatology, Johns Hopkins Medical Institutions, Baltimore, MD, USA; Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD, USA; Department of Oncology, Johns Hopkins Medical Institutions, Baltimore, MD, USA.
| |
Collapse
|
4229
|
Eggermont AM, Maio M, Robert C. Immune Checkpoint Inhibitors in Melanoma Provide the Cornerstones for Curative Therapies. Semin Oncol 2015; 42:429-35. [DOI: 10.1053/j.seminoncol.2015.02.010] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
4230
|
Klaver Y, Kunert A, Sleijfer S, Debets R, Lamers CHJ. Adoptive T-cell therapy: a need for standard immune monitoring. Immunotherapy 2015; 7:513-33. [DOI: 10.2217/imt.15.23] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Cancer immune therapy, in particular the use of checkpoint inhibitors and adoptive transfer of T cells has recently demonstrated significant clinical responses against several tumor types. Unfortunately, these therapies are frequently accompanied by severe toxicities, underscoring the need for markers that provide information on therapy response. Monitoring immune responses in the tumor microenvironment and peripheral blood prior to and during these therapies will provide better insight into the mechanisms underlying clinical activities, and will potentially enable the identification of such markers. In this review, we present an overview of adoptive T-cell trials conducted with a special focus on immune monitoring, and argue that accurate monitoring of T cells is pivotal to further development of immune therapies to treat cancer.
Collapse
Affiliation(s)
- Yarne Klaver
- Laboratory of Tumor Immunology, Department of Medical Oncology, Erasmus MC Cancer Institute, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Andre Kunert
- Laboratory of Tumor Immunology, Department of Medical Oncology, Erasmus MC Cancer Institute, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Stefan Sleijfer
- Laboratory of Tumor Immunology, Department of Medical Oncology, Erasmus MC Cancer Institute, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Reno Debets
- Laboratory of Tumor Immunology, Department of Medical Oncology, Erasmus MC Cancer Institute, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Cor HJ Lamers
- Laboratory of Tumor Immunology, Department of Medical Oncology, Erasmus MC Cancer Institute, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| |
Collapse
|
4231
|
Karachaliou N, Cao MG, Teixidó C, Viteri S, Morales-Espinosa D, Santarpia M, Rosell R. Understanding the function and dysfunction of the immune system in lung cancer: the role of immune checkpoints. Cancer Biol Med 2015; 12:79-86. [PMID: 26175923 PMCID: PMC4493378 DOI: 10.7497/j.issn.2095-3941.2015.0029] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 06/10/2015] [Indexed: 12/17/2022] Open
Abstract
Survival rates for metastatic lung cancer, including non-small cell lung cancer (NSCLC) and small cell lung cancer (SCLC), are poor with 5-year survivals of less than 5%. The immune system has an intricate and complex relationship with tumorigenesis; a groundswell of research on the immune system is leading to greater understanding of how cancer progresses and presenting new ways to halt disease progress. Due to the extraordinary power of the immune system-with its capacity for memory, exquisite specificity and central and universal role in human biology-immunotherapy has the potential to achieve complete, long-lasting remissions and cures, with few side effects for any cancer patient, regardless of cancer type. As a result, a range of cancer therapies are under development that work by turning our own immune cells against tumors. However deeper understanding of the complexity of immunomodulation by tumors is key to the development of effective immunotherapies, especially in lung cancer.
Collapse
Affiliation(s)
- Niki Karachaliou
- 1 Instituto Oncológico Dr Rosell, Quiron Dexeus University Hospital, Barcelona 08028, Spain ; 2 Pangaea Biotech, Barcelona 08028, Spain ; 3 Medical Oncology Unit, Human Pathology Department, University of Messina, Messina 98122, Italy ; 4 Catalan Institute of Oncology, Hospital Germans Trias i Pujol, Badalona 08916, Spain ; 5 Molecular Oncology Research (MORe) Foundation, Barcelona 08028, Spain ; 6 Germans Trias i Pujol Health Sciences Institute and Hospital, Campus Can Ruti 08916, Spain
| | - Maria Gonzalez Cao
- 1 Instituto Oncológico Dr Rosell, Quiron Dexeus University Hospital, Barcelona 08028, Spain ; 2 Pangaea Biotech, Barcelona 08028, Spain ; 3 Medical Oncology Unit, Human Pathology Department, University of Messina, Messina 98122, Italy ; 4 Catalan Institute of Oncology, Hospital Germans Trias i Pujol, Badalona 08916, Spain ; 5 Molecular Oncology Research (MORe) Foundation, Barcelona 08028, Spain ; 6 Germans Trias i Pujol Health Sciences Institute and Hospital, Campus Can Ruti 08916, Spain
| | - Cristina Teixidó
- 1 Instituto Oncológico Dr Rosell, Quiron Dexeus University Hospital, Barcelona 08028, Spain ; 2 Pangaea Biotech, Barcelona 08028, Spain ; 3 Medical Oncology Unit, Human Pathology Department, University of Messina, Messina 98122, Italy ; 4 Catalan Institute of Oncology, Hospital Germans Trias i Pujol, Badalona 08916, Spain ; 5 Molecular Oncology Research (MORe) Foundation, Barcelona 08028, Spain ; 6 Germans Trias i Pujol Health Sciences Institute and Hospital, Campus Can Ruti 08916, Spain
| | - Santiago Viteri
- 1 Instituto Oncológico Dr Rosell, Quiron Dexeus University Hospital, Barcelona 08028, Spain ; 2 Pangaea Biotech, Barcelona 08028, Spain ; 3 Medical Oncology Unit, Human Pathology Department, University of Messina, Messina 98122, Italy ; 4 Catalan Institute of Oncology, Hospital Germans Trias i Pujol, Badalona 08916, Spain ; 5 Molecular Oncology Research (MORe) Foundation, Barcelona 08028, Spain ; 6 Germans Trias i Pujol Health Sciences Institute and Hospital, Campus Can Ruti 08916, Spain
| | - Daniela Morales-Espinosa
- 1 Instituto Oncológico Dr Rosell, Quiron Dexeus University Hospital, Barcelona 08028, Spain ; 2 Pangaea Biotech, Barcelona 08028, Spain ; 3 Medical Oncology Unit, Human Pathology Department, University of Messina, Messina 98122, Italy ; 4 Catalan Institute of Oncology, Hospital Germans Trias i Pujol, Badalona 08916, Spain ; 5 Molecular Oncology Research (MORe) Foundation, Barcelona 08028, Spain ; 6 Germans Trias i Pujol Health Sciences Institute and Hospital, Campus Can Ruti 08916, Spain
| | - Mariacarmela Santarpia
- 1 Instituto Oncológico Dr Rosell, Quiron Dexeus University Hospital, Barcelona 08028, Spain ; 2 Pangaea Biotech, Barcelona 08028, Spain ; 3 Medical Oncology Unit, Human Pathology Department, University of Messina, Messina 98122, Italy ; 4 Catalan Institute of Oncology, Hospital Germans Trias i Pujol, Badalona 08916, Spain ; 5 Molecular Oncology Research (MORe) Foundation, Barcelona 08028, Spain ; 6 Germans Trias i Pujol Health Sciences Institute and Hospital, Campus Can Ruti 08916, Spain
| | - Rafael Rosell
- 1 Instituto Oncológico Dr Rosell, Quiron Dexeus University Hospital, Barcelona 08028, Spain ; 2 Pangaea Biotech, Barcelona 08028, Spain ; 3 Medical Oncology Unit, Human Pathology Department, University of Messina, Messina 98122, Italy ; 4 Catalan Institute of Oncology, Hospital Germans Trias i Pujol, Badalona 08916, Spain ; 5 Molecular Oncology Research (MORe) Foundation, Barcelona 08028, Spain ; 6 Germans Trias i Pujol Health Sciences Institute and Hospital, Campus Can Ruti 08916, Spain
| |
Collapse
|
4232
|
Sullivan RJ, Flaherty KT. New Strategies in Melanoma: Entering the Era of Combinatorial Therapy. Clin Cancer Res 2015; 21:2424-35. [DOI: 10.1158/1078-0432.ccr-14-1650] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
4233
|
Cherny NI, Sullivan R, Dafni U, Kerst JM, Sobrero A, Zielinski C, de Vries EGE, Piccart MJ. A standardised, generic, validated approach to stratify the magnitude of clinical benefit that can be anticipated from anti-cancer therapies: the European Society for Medical Oncology Magnitude of Clinical Benefit Scale (ESMO-MCBS). Ann Oncol 2015; 26:1547-73. [PMID: 26026162 DOI: 10.1093/annonc/mdv249] [Citation(s) in RCA: 621] [Impact Index Per Article: 62.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 05/22/2015] [Indexed: 12/12/2022] Open
Abstract
The value of any new therapeutic strategy or treatment is determined by the magnitude of its clinical benefit balanced against its cost. Evidence for clinical benefit from new treatment options is derived from clinical research, in particular phase III randomised trials, which generate unbiased data regarding the efficacy, benefit and safety of new therapeutic approaches. To date, there is no standard tool for grading the magnitude of clinical benefit of cancer therapies, which may range from trivial (median progression-free survival advantage of only a few weeks) to substantial (improved long-term survival). Indeed, in the absence of a standardised approach for grading the magnitude of clinical benefit, conclusions and recommendations derived from studies are often hotly disputed and very modest incremental advances have often been presented, discussed and promoted as major advances or 'breakthroughs'. Recognising the importance of presenting clear and unbiased statements regarding the magnitude of the clinical benefit from new therapeutic approaches derived from high-quality clinical trials, the European Society for Medical Oncology (ESMO) has developed a validated and reproducible tool to assess the magnitude of clinical benefit for cancer medicines, the ESMO Magnitude of Clinical Benefit Scale (ESMO-MCBS). This tool uses a rational, structured and consistent approach to derive a relative ranking of the magnitude of clinically meaningful benefit that can be expected from a new anti-cancer treatment. The ESMO-MCBS is an important first step to the critical public policy issue of value in cancer care, helping to frame the appropriate use of limited public and personal resources to deliver cost-effective and affordable cancer care. The ESMO-MCBS will be a dynamic tool and its criteria will be revised on a regular basis.
Collapse
Affiliation(s)
- N I Cherny
- Cancer Pain and Palliative Medicine Service, Department of Medical Oncology, Shaare Zedek Medical Center, Jerusalem, Israel
| | - R Sullivan
- Kings Health Partners Integrated Cancer Centre, King's College London, Institute of Cancer Policy, London, UK
| | - U Dafni
- University of Athens and Frontiers of Science Foundation-Hellas, Athens, Greece
| | - J M Kerst
- Department of Medical Oncology, Antoni van Leeuwenhoek Hospital
| | - A Sobrero
- Department of Medical Oncology, IRCCS San Martino IST, Genova, Italy
| | - C Zielinski
- Division of Oncology, Medical University Vienna, Vienna, Austria
| | - E G E de Vries
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - M J Piccart
- Jules Bordet Institute, UniversitéLibre de Bruxelles, Brussels, Belgium Netherlands Cancer Institute, Amsterdam, The Netherlands
| |
Collapse
|
4234
|
Zhang Y, Song Y, Gao Q. Increased survival time of a patient with metastatic malignant melanoma following immunotherapy: A case report and literature review. Oncol Lett 2015; 10:883-886. [PMID: 26622588 DOI: 10.3892/ol.2015.3296] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 04/13/2015] [Indexed: 01/02/2023] Open
Abstract
Metastatic malignant melanoma is treated with chemotherapy and radiotherapy. A number of previous studies have indicated that cytokine-induced killer cells (CIK cells) are a heterogeneous cell population that express cluster of differentiation (CD)3 and CD56, in addition to the natural killer cell NKG2D activating receptor. CIK cells possess major histocompatibility complex-unrestricted cytotoxicity towards cancer, but not towards normal targets. The present study investigated whether the addition of CIK cells resulted in an improved therapeutic response in a patient with metastatic malignant melanoma. In the current case, a patient with metastatic malignant melanoma received CIK therapy, which resulted in a relatively long survival time of 28 months. To the best of our knowledge, there have been no previous studies reporting such positive effects in a patient who received CIK cell immunotherapy. Based on the findings of the present study, CIK cell therapy may be an option that results in a good prognosis in certain patients with metastatic malignant melanoma.
Collapse
Affiliation(s)
- Yong Zhang
- Department of Immunotherapy, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan 450003, P.R. China
| | - Yongping Song
- Department of Immunotherapy, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan 450003, P.R. China
| | - Quanli Gao
- Department of Immunotherapy, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan 450003, P.R. China
| |
Collapse
|
4235
|
Sundarasetty BS, Chan L, Darling D, Giunti G, Farzaneh F, Schenck F, Naundorf S, Kuehlcke K, Ruggiero E, Schmidt M, von Kalle C, Rothe M, Hoon DSB, Gerasch L, Figueiredo C, Koehl U, Blasczyk R, Gutzmer R, Stripecke R. Lentivirus-induced 'Smart' dendritic cells: Pharmacodynamics and GMP-compliant production for immunotherapy against TRP2-positive melanoma. Gene Ther 2015; 22:707-20. [PMID: 25965393 PMCID: PMC4561294 DOI: 10.1038/gt.2015.43] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Accepted: 03/23/2015] [Indexed: 02/06/2023]
Abstract
Monocyte-derived conventional dendritic cells (ConvDCs) loaded with melanoma antigens showed modest responses in clinical trials. Efficacy studies were hampered by difficulties in ConvDC manufacturing and low potency. Overcoming these issues, we demonstrated higher potency of lentiviral vector (LV)-programmed DCs. Monocytes were directly induced to self-differentiate into DCs (SmartDC-TRP2) upon transduction with a tricistronic LV encoding for cytokines (granulocyte macrophage colony stimulating factor (GM-CSF) and interleukin-4 (IL-4)) and a melanoma antigen (tyrosinase-related protein 2 (TRP2)). Here, SmartDC-TRP2 generated with monocytes from five advanced melanoma patients were tested in autologous DC:T cell stimulation assays, validating the activation of functional TRP2-specific cytotoxic T lymphocytes (CTLs) for all patients. We described methods compliant to good manufacturing practices (GMP) to produce LV and SmartDC-TRP2. Feasibility of monocyte transduction in a bag system and cryopreservation following a 24-h standard operating procedure were achieved. After thawing, 50% of the initial monocyte input was recovered and SmartDC-TRP2 self-differentiated in vitro, showing uniform expression of DC markers, detectable LV copies and a polyclonal LV integration pattern not biased to oncogenic loci. GMP-grade SmartDC-TRP2 expanded TRP2-specific autologous CTLs in vitro. These results demonstrated a simpler GMP-compliant method of manufacturing an effective individualized DC vaccine. Such DC vaccine, when in combination with checkpoint inhibition therapies, might provide higher specificity against melanoma.
Collapse
Affiliation(s)
- B S Sundarasetty
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - L Chan
- Department of Hematological Medicine, Cell and Gene Therapy at King's, The Rayne Institute, King's College London, London, UK
| | - D Darling
- Department of Hematological Medicine, Cell and Gene Therapy at King's, The Rayne Institute, King's College London, London, UK
| | - G Giunti
- Department of Hematological Medicine, Cell and Gene Therapy at King's, The Rayne Institute, King's College London, London, UK
| | - F Farzaneh
- Department of Hematological Medicine, Cell and Gene Therapy at King's, The Rayne Institute, King's College London, London, UK
| | - F Schenck
- Department of Dermatology and Allergy, Skin Cancer Center Hannover, Hannover Medical School, Hannover, Germany
| | - S Naundorf
- EUFETS GmbH, Idar-Oberstein, Heidelberg, Germany
| | - K Kuehlcke
- EUFETS GmbH, Idar-Oberstein, Heidelberg, Germany
| | - E Ruggiero
- Division of Translational Oncology, National Center for Tumor Diseases, Heidelberg, Germany
| | - M Schmidt
- Division of Translational Oncology, National Center for Tumor Diseases, Heidelberg, Germany
| | - C von Kalle
- Division of Translational Oncology, National Center for Tumor Diseases, Heidelberg, Germany
| | - M Rothe
- Department of Experimental Hematology, Hannover, Germany
| | - D S B Hoon
- John Wayne Cancer Institute, Santa Monica, CA, USA
| | - L Gerasch
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - C Figueiredo
- Department of Transfusion Medicine, Hannover Medical School, Hannover, Germany
| | - U Koehl
- Institute for Cell Therapeutics and GMP core facility IFB-Tx, Hannover Medical School, Hannover, Germany
| | - R Blasczyk
- Department of Transfusion Medicine, Hannover Medical School, Hannover, Germany
| | - R Gutzmer
- Department of Dermatology and Allergy, Skin Cancer Center Hannover, Hannover Medical School, Hannover, Germany
| | - R Stripecke
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| |
Collapse
|
4236
|
Massey PR, Prasad V, Figg WD, Fojo T. Multiplying therapies and reducing toxicity in metastatic melanoma. Cancer Biol Ther 2015; 16:1014-8. [PMID: 26016850 DOI: 10.1080/15384047.2015.1046650] [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: 10/23/2022] Open
Abstract
Prior to 2011, only 2 systemic treatments were approved for the treatment of melanoma and these had limited efficacy. In the past 4 years, 6 novel agents have received FDA approval. Herein, we will focus on 4 recently published NEJM papers reporting the results of clinical trials, comprising 4 agents targeting the MAPK pathway: the BRAF inhibitors vemurafenib and dabrafenib, and the MEK inhibitors trametinib and cobimetenib. These have been developed in parallel with a class of immunologic mediators often referred to as "immune checkpoint inhibitors." These recent studies represent a marked acceleration of progress in the treatment of metastatic melanoma. While it was hoped that combining BRAF and MEK inhibitors would significantly mitigate drug resistance, such combinations have yielded only modestly better results than monotherapy. However, these combinations were successful in reducing the development of cutaneous squamous cell carcinomas and keratocanthomas. Therefore, combination therapies are clearly warranted. Thus far there are only limited data addressing the value of combinations of immunotherapeutic agents: a phase 1 trial of concurrent nivolumab plus ipilimumab suggested enhanced activity that may not depend on BRAF mutation status. Despite the attention and publicity given to the progress achieved in the therapy of melanoma, the majority of patients with metastatic disease still have a poor prognosis. Even novel combination regiments of BRAF and MEK inhibitors achieve complete response in only 13% of patients and a median PFS of 11.4 months in all patients. Better therapies remain desperately needed, especially for the 30-40% of patients with wild-type BRAF, for whom BRAF/MAPK inhibition offers no benefit. In the latter benefit is expected from emerging immunotherapies either singly or in combinations. The extent to which immunotherapies will add to regimens targeting BRAF remains to be determined.
Collapse
Affiliation(s)
- Paul R Massey
- a Dell Medical School; The University of Texas at Austin ; Austin , TX , USA
| | | | | | | |
Collapse
|
4237
|
Affiliation(s)
- Dario Neri
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH Zürich), Zürich, Switzerland
| |
Collapse
|
4238
|
Abstract
Cancer is a close second to heart disease for cause of death in the USA, and could soon surpass heart disease as the population ages and the incidence of cancer continues to increase. While heart disease can be addressed through behavior modification and education (e.g., smoking cessation, dietary changes, exercises that promote cardiovascular fitness), pharmacology and improved surgical devices and methods, cancer ultimately requires improved and novel drug treatments to bring mortality rates down. In 2014, the US FDA approved 17 drugs and/or drug combinations in 12 disease sites for a total of 19 indications in melanoma, hematologic malignancies, gastrointestinal carcinoma, non-small-cell lung cancer, gynecologic malignancies and lymphoma/lymphoproliferative disorders.
Collapse
Affiliation(s)
- Juliet E Wolford
- The Chao Family NCI-Designated Comprehensive Cancer Center, University of California, Irvine Medical Center, 101 The City Drive South, Bldg 56, Orange, CA 92868, USA
| | - Krishnansu S Tewari
- The Chao Family NCI-Designated Comprehensive Cancer Center, University of California, Irvine Medical Center, 101 The City Drive South, Bldg 56, Orange, CA 92868, USA
| |
Collapse
|
4239
|
Postow MA, Chesney J, Pavlick AC, Robert C, Grossmann K, McDermott D, Linette GP, Meyer N, Giguere JK, Agarwala SS, Shaheen M, Ernstoff MS, Minor D, Salama AK, Taylor M, Ott PA, Rollin LM, Horak C, Gagnier P, Wolchok JD, Hodi FS. Nivolumab and ipilimumab versus ipilimumab in untreated melanoma. N Engl J Med 2015; 372:2006-17. [PMID: 25891304 PMCID: PMC5744258 DOI: 10.1056/nejmoa1414428] [Citation(s) in RCA: 2208] [Impact Index Per Article: 220.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND In a phase 1 dose-escalation study, combined inhibition of T-cell checkpoint pathways by nivolumab and ipilimumab was associated with a high rate of objective response, including complete responses, among patients with advanced melanoma. METHODS In this double-blind study involving 142 patients with metastatic melanoma who had not previously received treatment, we randomly assigned patients in a 2:1 ratio to receive ipilimumab (3 mg per kilogram of body weight) combined with either nivolumab (1 mg per kilogram) or placebo once every 3 weeks for four doses, followed by nivolumab (3 mg per kilogram) or placebo every 2 weeks until the occurrence of disease progression or unacceptable toxic effects. The primary end point was the rate of investigator-assessed, confirmed objective response among patients with BRAF V600 wild-type tumors. RESULTS Among patients with BRAF wild-type tumors, the rate of confirmed objective response was 61% (44 of 72 patients) in the group that received both ipilimumab and nivolumab (combination group) versus 11% (4 of 37 patients) in the group that received ipilimumab and placebo (ipilimumab-monotherapy group) (P<0.001), with complete responses reported in 16 patients (22%) in the combination group and no patients in the ipilimumab-monotherapy group. The median duration of response was not reached in either group. The median progression-free survival was not reached with the combination therapy and was 4.4 months with ipilimumab monotherapy (hazard ratio associated with combination therapy as compared with ipilimumab monotherapy for disease progression or death, 0.40; 95% confidence interval, 0.23 to 0.68; P<0.001). Similar results for response rate and progression-free survival were observed in 33 patients with BRAF mutation-positive tumors. Drug-related adverse events of grade 3 or 4 were reported in 54% of the patients who received the combination therapy as compared with 24% of the patients who received ipilimumab monotherapy. Select adverse events with potential immunologic causes were consistent with those in a phase 1 study, and most of these events resolved with immune-modulating medication. CONCLUSIONS The objective-response rate and the progression-free survival among patients with advanced melanoma who had not previously received treatment were significantly greater with nivolumab combined with ipilimumab than with ipilimumab monotherapy. Combination therapy had an acceptable safety profile. (Funded by Bristol-Myers Squibb; ClinicalTrials.gov number, NCT01927419.).
Collapse
Affiliation(s)
- Michael A. Postow
- Memorial Sloan Kettering Cancer Center, New York, NY
- Weill Cornell Medical College, New York, NY
| | - Jason Chesney
- J. Graham Brown Cancer Center, University of Louisville, Louisville, KY
| | | | - Caroline Robert
- Gustave, Roussy and Paris-Sud University, Villejuif-Paris-Sud, France
| | | | | | | | | | | | | | | | - Marc S. Ernstoff
- Taussig Cancer Institute, Cleveland Clinic Foundation, Cleveland, OH
| | - David Minor
- California Pacific Center for Melanoma Research, San Francisco, CA
| | | | | | | | | | | | | | | | | |
Collapse
|
4240
|
Chapman PB, D'Angelo SP, Wolchok JD. Rapid eradication of a bulky melanoma mass with one dose of immunotherapy. N Engl J Med 2015; 372:2073-4. [PMID: 25891305 DOI: 10.1056/nejmc1501894] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
4241
|
Barbee MS, Ogunniyi A, Horvat TZ, Dang TO. Current status and future directions of the immune checkpoint inhibitors ipilimumab, pembrolizumab, and nivolumab in oncology. Ann Pharmacother 2015; 49:907-37. [PMID: 25991832 DOI: 10.1177/1060028015586218] [Citation(s) in RCA: 140] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
OBJECTIVE To provide the clinician with an update and the current status and future direction of approved immune checkpoint inhibitors (ICIs) in oncology. DATA SOURCES A PubMed search from January 1, 1966 to March 13, 2015 was performed using the key terms ipilimumab, pembrolizumab, lambrolizumab, nivolumab, immune checkpoint inhibitor, MDX-010, MDX-101, BMS-734016, MK-3475, SCH 900475, MDX-1106, BMS-936558, ONO-4538, CTLA-4, PD-1, or PD-L1 and cancer, oncology, or neoplasm. Additional references were identified from the investigators(') personal files, recent oncology meetings, review articles, clinical guidelines, and package inserts. STUDY SELECTION AND DATA EXTRACTION All English-language clinical trials assessing the safety and efficacy of ipilimumab, nivolumab, and pembrolizumab in cancer were considered. The PubMed search resulted in 215 trials; 33 met inclusion criteria. A further 28 trials were identified from the above sources; 61 trials from 2005 to 2015 were included. We consolidated and clarified treatment recommendations for the management of immune-related adverse events (irAEs), assessed response criteria, and calculated the clinical utility of leading tumor profiling options. DATA SYNTHESIS Ipilimumab and nivolumab, but not pembrolizumab, have an overall survival (OS) advantage over chemotherapy first line in unresectable/metastatic melanoma. Nivolumab has an OS advantage versus chemotherapy in second-line squamous non-small-cell lung cancer. Data in other settings are promising. Nivolumab and pembrolizumab are better tolerated than ipilimumab. Further validation of response criteria is needed. Tumor profiling to predict clinical benefit is premature but promising. CONCLUSIONS The treatment landscape in oncology is quickly evolving with the advent of ICIs.
Collapse
Affiliation(s)
| | | | - Troy Z Horvat
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Thu-Oanh Dang
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
4242
|
Ding X, Cao H, Chen X, Jin H, Liu Z, Wang G, Cai L, Li D, Niu C, Tian H, Yang L, Zhao Y, Li W, Cui J. Cellular immunotherapy as maintenance therapy prolongs the survival of the patients with small cell lung cancer. J Transl Med 2015; 13:158. [PMID: 25968637 PMCID: PMC4446113 DOI: 10.1186/s12967-015-0514-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 05/04/2015] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Small cell lung cancer (SCLC) relapses rapidly after the initial response to chemotherapy and shows drug-resistance. This study was to investigate the efficacy and safety of cellular immunotherapy (CIT) with autologous natural killer (NK), γδT, and cytokine-induced killer (CIK) cells as maintenance therapy for SCLC patients. METHODS A pilot prospective cohort study was conducted with SCLC patients who had responded to initial chemotherapy. Patients elected to receive either CIT as maintenance therapy (study group), or to be followed-up without further treatment (control group). Progression-free survival (PFS), overall survival (OS), and adverse effects were investigated. RESULTS We recruited 58 patients (29 in each group). The patient characteristics of the 2 groups were well balanced. PFS was not significantly different between the groups, but OS was significantly longer in the study group than the control (20 vs. 11.5 months, P = 0.005; hazard ratio [HR], 0.434, 95 % confidence interval [CI], 0.236-0.797, P = 0.007). Among patients with limited-stage disease, there was no difference in PFS between the groups, but OS was longer in the study group compared to the control (26.5 vs. 11.8 months, P = 0.033; HR, 0.405, 95 % CI, 0.169-0.972, P = 0.043). Among patients with extensive-stage disease, both PFS and OS were longer in the study group than the control (5 vs. 2.7 months, P = 0.037; HR, 0.403, 95 % CI, 0.162-1.003, P = 0.051, and 14.5 vs. 9 months, P = 0.038; HR, 0.403, 95 % CI, 0.165-0.987, P = 0.047, respectively). No significant adverse reactions occurred in patients undergoing CIT. CONCLUSIONS CIT maintenance therapy in SCLC prolonged survival with only minimal side effects. Integrating CIT into current treatment may be a novel strategy for SCLC therapy, although further multi-center randomized studies are needed.
Collapse
Affiliation(s)
- Xiao Ding
- Cancer Center, the First Hospital of Jilin University, No. 71. Xinmin Street, Changchun, 130021, China.
| | - He Cao
- Cancer Center, the First Hospital of Jilin University, No. 71. Xinmin Street, Changchun, 130021, China.
| | - Xiao Chen
- Cancer Center, the First Hospital of Jilin University, No. 71. Xinmin Street, Changchun, 130021, China.
| | - Haofan Jin
- Cancer Center, the First Hospital of Jilin University, No. 71. Xinmin Street, Changchun, 130021, China.
| | - Ziling Liu
- Cancer Center, the First Hospital of Jilin University, No. 71. Xinmin Street, Changchun, 130021, China.
| | - Guanjun Wang
- Cancer Center, the First Hospital of Jilin University, No. 71. Xinmin Street, Changchun, 130021, China.
| | - Lu Cai
- Kosair Children's Hospital Research Institute, Department of Pediatrics, the University of Louisville, Louisville, KY, 40202, USA.
| | - Dan Li
- Cancer Center, the First Hospital of Jilin University, No. 71. Xinmin Street, Changchun, 130021, China.
| | - Chao Niu
- Cancer Center, the First Hospital of Jilin University, No. 71. Xinmin Street, Changchun, 130021, China.
| | - Huimin Tian
- Cancer Center, the First Hospital of Jilin University, No. 71. Xinmin Street, Changchun, 130021, China.
| | - Lei Yang
- Cancer Center, the First Hospital of Jilin University, No. 71. Xinmin Street, Changchun, 130021, China.
| | - Yuguang Zhao
- Cancer Center, the First Hospital of Jilin University, No. 71. Xinmin Street, Changchun, 130021, China.
| | - Wei Li
- Cancer Center, the First Hospital of Jilin University, No. 71. Xinmin Street, Changchun, 130021, China.
| | - Jiuwei Cui
- Cancer Center, the First Hospital of Jilin University, No. 71. Xinmin Street, Changchun, 130021, China.
| |
Collapse
|
4243
|
Gibney GT, Atkins MB. Swinging for the Fences: Long-Term Survival With Ipilimumab in Metastatic Melanoma. J Clin Oncol 2015; 33:1873-7. [PMID: 25964248 DOI: 10.1200/jco.2014.60.1807] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
A 40-year-old man with stage III melanoma arising from his left shoulder underwent wide local excision, sentinel lymph node biopsy, and lymph node dissection. Nine months after receiving adjuvant biochemotherapy with cisplatin, vinblastine, dacarbazine, interleukin-2 (IL-2), and interferon alfa as part of a clinical trial, he developed headaches and right-hand weakness and was found to have a 2-cm left parietal CNS metastasis. A comprehensive staging workup identified multiple nonspecific subcentimeter pulmonary nodules. The brain mass was resected and confirmed to be metastatic melanoma; the surgical bed was treated with stereotactic radiosurgery. He was monitored off therapy, but 5 months later, he developed a second left parietal CNS metastasis and enlarging lung nodules. The new brain lesion was treated with stereotactic radiosurgery, and he began systemic therapy with ipilimumab on a clinical trial. After the third dose, he presented with headache, nausea, and vomiting; a brain magnetic resonance imaging scan showed left anterior temporal enhancement, possibly representing new disease. His symptoms improved with a course of corticosteroids. Restaging of the chest showed a mixed response among the pulmonary nodules. After tapering off corticosteroids, he received the fourth dose of ipilimumab, which was complicated by grade 3 transaminitis and hypophysitis with documented hypothyroidism and adrenal insufficiency. They were managed with corticosteroids and thyroid and adrenal hormone replacement. Restaging scans showed further disease regression except for new confluent enhancing nodules and edema in the left temporal lobe. Craniotomy and resection of this area showed only necrotic tissue with no viable melanoma cells. Nine years after treatment with ipilimumab, he is alive and shows no evidence of melanoma on the basis of annual computed tomography scans of the chest, abdomen, and pelvis and magnetic resonance imaging scans of the brain. He has full neurologic function but still requires hormone replacement for persistent hypopituitarism.
Collapse
Affiliation(s)
| | - Michael B Atkins
- Georgetown-Lombardi Comprehensive Cancer Center; and Medstar-Georgetown University Hospital, Washington, DC
| |
Collapse
|
4244
|
Nsengimana J, Laye J, Filia A, Walker C, Jewell R, Van den Oord JJ, Wolter P, Patel P, Sucker A, Schadendorf D, Jönsson GB, Bishop DT, Newton-Bishop J. Independent replication of a melanoma subtype gene signature and evaluation of its prognostic value and biological correlates in a population cohort. Oncotarget 2015; 6:11683-93. [PMID: 25871393 PMCID: PMC4484486 DOI: 10.18632/oncotarget.3549] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Accepted: 02/10/2015] [Indexed: 12/05/2022] Open
Abstract
Development and validation of robust molecular biomarkers has so far been limited in melanoma research. In this paper we used a large population-based cohort to replicate two published gene signatures for melanoma classification. We assessed the signatures prognostic value and explored their biological significance by correlating them with factors known to be associated with survival (vitamin D) or etiological routes (nevi, sun sensitivity and telomere length). Genomewide microarray gene expressions were profiled in 300 archived tumors (224 primaries, 76 secondaries). The two gene signatures classified up to 96% of our samples and showed strong correlation with melanoma specific survival (P=3 x 10(-4)), Breslow thickness (P=5 x 10(-10)), ulceration (P=9.x10-8) and mitotic rate (P=3 x 10(-7)), adding prognostic value over AJCC stage (adjusted hazard ratio 1.79, 95%CI 1.13-2.83), as previously reported. Furthermore, molecular subtypes were associated with season-adjusted serum vitamin D at diagnosis (P=0.04) and genetically predicted telomere length (P=0.03). Specifically, molecular high-grade tumors were more frequent in patients with lower vitamin D levels whereas high immune tumors came from patients with predicted shorter telomeres. Our data confirm the utility of molecular biomarkers in melanoma prognostic estimation using tiny archived specimens and shed light on biological mechanisms likely to impact on cancer initiation and progression.
Collapse
Affiliation(s)
- Jérémie Nsengimana
- Section of Epidemiology and Biostatistics, Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, UK
| | - Jon Laye
- Section of Epidemiology and Biostatistics, Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, UK
| | - Anastasia Filia
- National Heart and Lung Institute, Imperial College, London, UK
| | - Christy Walker
- Section of Epidemiology and Biostatistics, Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, UK
| | - Rosalyn Jewell
- Yorkshire Regional Genetics Service, Chapel Allerton Hospital, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Joost J Van den Oord
- Department of Pathology, University Hospitals Leuven, Leuven, Belgium
- European Organisation for Research and Treatment of Cancer (EORTC) Melanoma Group, Brussels, Belgium
| | - Pascal Wolter
- Department of General Medical Oncology, Leuven Cancer Institute, University Hospitals Leuven, Leuven, Belgium
| | - Poulam Patel
- School of Medicine, University of Nottingham, Nottingham, UK
- European Organisation for Research and Treatment of Cancer (EORTC) Melanoma Group, Brussels, Belgium
| | - Antje Sucker
- Department of Dermatology, Essen University Hospital, Essen, and German Consortium of Translational Cancer Research (DKTK), Heidelberg, Germany
| | - Dirk Schadendorf
- Department of Dermatology, Essen University Hospital, Essen, and German Consortium of Translational Cancer Research (DKTK), Heidelberg, Germany
- European Organisation for Research and Treatment of Cancer (EORTC) Melanoma Group, Brussels, Belgium
| | - Göran B Jönsson
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - D. Timothy Bishop
- Section of Epidemiology and Biostatistics, Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, UK
| | - Julia Newton-Bishop
- Section of Epidemiology and Biostatistics, Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, UK
| |
Collapse
|
4245
|
Firor AE, Jares A, Ma Y. From humble beginnings to success in the clinic: Chimeric antigen receptor-modified T-cells and implications for immunotherapy. Exp Biol Med (Maywood) 2015; 240:1087-98. [PMID: 25956686 DOI: 10.1177/1535370215584936] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 03/31/2015] [Indexed: 11/15/2022] Open
Abstract
In the past 50 years, disease burden has steadily shifted from infectious disease to cancer. Standard chemotherapy has long been the mainstay of cancer medical management, and despite vast efforts towards more targeted and personalized drug therapy, many cancers remain refractory to treatment, with high rates of relapse and poor prognosis. Recent dramatic immunotherapy clinical trials have demonstrated that engineering T-cells with chimeric antigen receptors (CARs) to target CD19 can lead to complete remission in relapsed or refractory B-cell malignancies, generating a great deal of enthusiasm in the field. Here we provide a comprehensive overview of the history of adoptive T-cell therapy, including CARs, in solid tumors as well as hematologic malignancies. CAR therapy has the potential to fundamentally transform cancer treatment with specific and even personalized targeting of tissue- and tumor-specific antigens. However, before CARs become standard first-line treatment modalities, critical issues regarding efficacy, combinatorial regimens, and mechanisms of treatment failure and toxicity will need to be addressed.
Collapse
Affiliation(s)
- Amelia E Firor
- Department of Pathology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Alexander Jares
- Department of Pathology, Stony Brook University, Stony Brook, NY 11794, USA Medical Scientist Training Program, Stony Brook University, Stony Brook, NY 11794, USA Graduate Program in Molecular and Cellular Pharmacology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Yupo Ma
- Department of Pathology, Stony Brook University, Stony Brook, NY 11794, USA
| |
Collapse
|
4246
|
Taube JM, Young GD, McMiller TL, Chen S, Salas JT, Pritchard TS, Xu H, Meeker AK, Fan J, Cheadle C, Berger AE, Pardoll DM, Topalian SL. Differential Expression of Immune-Regulatory Genes Associated with PD-L1 Display in Melanoma: Implications for PD-1 Pathway Blockade. Clin Cancer Res 2015; 21:3969-76. [PMID: 25944800 DOI: 10.1158/1078-0432.ccr-15-0244] [Citation(s) in RCA: 202] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Accepted: 04/16/2015] [Indexed: 12/13/2022]
Abstract
PURPOSE Blocking the immunosuppressive PD-1/PD-L1 pathway has antitumor activity in multiple cancer types, and PD-L1 expression on tumor cells and infiltrating myeloid cells correlates with the likelihood of response. We previously found that IFNG (interferon-gamma) was overexpressed by tumor-infiltrating lymphocytes in PD-L1(+) versus PD-L1(-) melanomas, creating adaptive immune resistance by promoting PD-L1 display. This study was undertaken to identify additional factors in the PD-L1(+) melanoma microenvironment coordinately contributing to immunosuppression. EXPERIMENTAL DESIGN Archived, formalin-fixed paraffin-embedded melanoma specimens were assessed for PD-L1 protein expression at the tumor cell surface with IHC. Whole-genome expression analysis, quantitative (q)RT-PCR, IHC, and functional in vitro validation studies were used to assess factors differentially expressed in PD-L1(+) versus PD-L1(-) melanomas. RESULTS Functional annotation clustering based on whole-genome expression profiling revealed pathways upregulated in PD-L1(+) melanomas, involving immune cell activation, inflammation, and antigen processing and presentation. Analysis by qRT-PCR demonstrated overexpression of functionally related genes in PD-L1(+) melanomas, involved in CD8(+) T-cell activation (CD8A, IFNG, PRF1, and CCL5), antigen presentation (CD163, TLR3, CXCL1, and LYZ), and immunosuppression [PDCD1 (PD-1), CD274 (PD-L1), and LAG3, IL10]. Functional studies demonstrated that some factors, including IL10 and IL32-gamma, induced PD-L1 expression on monocytes but not tumor cells. CONCLUSIONS These studies elucidate the complexity of immune checkpoint regulation in the tumor microenvironment, identifying multiple factors likely contributing to coordinated immunosuppression. These factors may provide tumor escape mechanisms from anti-PD-1/PD-L1 therapy, and should be considered for cotargeting in combinatorial immunomodulation treatment strategies.
Collapse
Affiliation(s)
- Janis M Taube
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland. Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland. Department of Oncology, Sidney Kimmel Comprehensive Cancer Center and Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Geoffrey D Young
- Department of Otolaryngology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Tracee L McMiller
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Shuming Chen
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - January T Salas
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Theresa S Pritchard
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Haiying Xu
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Alan K Meeker
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jinshui Fan
- The Lowe Family Genomics Core, Sidney Kimmel Comprehensive Cancer Center and Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Chris Cheadle
- The Lowe Family Genomics Core, Sidney Kimmel Comprehensive Cancer Center and Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Alan E Berger
- The Lowe Family Genomics Core, Sidney Kimmel Comprehensive Cancer Center and Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Drew M Pardoll
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center and Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Suzanne L Topalian
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| |
Collapse
|
4247
|
Abstract
The complexity of cancer chemotherapy requires pharmacists be familiar with the complicated regimens and highly toxic agents used. This column reviews various issues related to preparation, dispensing, and administration of antineoplastic therapy, and the agents, both commercially available and investigational, used to treat malignant diseases. Questions or suggestions for topics should be addressed to Dominic A. Solimando, Jr, President, Oncology Pharmacy Services, Inc., 4201 Wilson Blvd #110-545, Arlington, VA 22203, e-mail: OncRxSvc@comcast.net; or J. Aubrey Waddell, Professor, University of Tennessee College of Pharmacy; Oncology Pharmacist, Pharmacy Department, Blount Memorial Hospital, 907 E. Lamar Alexander Parkway, Maryville, TN 37804, e-mail: waddfour@charter.net.
Collapse
|
4248
|
Brahmer JR, Hammers H, Lipson EJ. Nivolumab: targeting PD-1 to bolster antitumor immunity. Future Oncol 2015; 11:1307-26. [DOI: 10.2217/fon.15.52] [Citation(s) in RCA: 141] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
4249
|
Prise en charge des carcinomes épidermoïdes de la tête et du cou à la phase métastatique. ONCOLOGIE 2015. [DOI: 10.1007/s10269-015-2521-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
4250
|
Giannini I, Cutrignelli DA, Resta L, Gentile A, Vincenti L. Metastatic melanoma of the gallbladder: report of two cases and a review of the literature. Clin Exp Med 2015; 16:295-300. [PMID: 25929736 DOI: 10.1007/s10238-015-0353-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 04/20/2015] [Indexed: 12/14/2022]
Abstract
Melanoma is one of the most aggressive and highly metastatic cancers. The most common sites of distant metastases are soft tissues, lung, liver, skin and brain, whereas only few patients develop gastrointestinal metastases. Metastatic involvement of the gallbladder is rare and more often part of a widespread disease than a solitary lesion. The "gold-standard" treatment of metastatic melanoma of the gallbladder remains unclear. We report two cases of patients with past history of cutaneous melanoma who developed visceral metastases. The first patient was asymptomatic and had a widespread disease with metastatic involvement of both the spleen and the gallbladder. The second patient had an isolated metastasis of the gallbladder and complained of upper abdominal pain. The chosen treatment was open cholecystectomy (and splenectomy) in the first case and laparoscopic cholecystectomy in the second. A review of the literature is provided.
Collapse
Affiliation(s)
- I Giannini
- General Surgery and Liver Transplantation Unit, Department of Emergency and Organ Transplantation, University "A. Moro" of Bari, Policlinico. Piazza G. Cesare, 11, 70124, Bari, Italy.
| | - D A Cutrignelli
- Complex Operating Unit of Plastic Aesthetic and Reconstructive Surgery, Department of Emergency and Organ Transplantation, University "A. Moro" of Bari, Bari, Italy
| | - L Resta
- Department of Emergence and Organ Transplantation, Institute of Pathological Anatomy, University "A. Moro" of Bari, Bari, Italy
| | - A Gentile
- Department of Emergence and Organ Transplantation, Institute of Pathological Anatomy, University "A. Moro" of Bari, Bari, Italy
| | - L Vincenti
- General Surgery Unit, Policlinico of Bari, Bari, Italy
| |
Collapse
|