4251
|
Mackiewicz J, Karczewska-Dzionk A, Laciak M, Kapcinska M, Wiznerowicz M, Burzykowski T, Zakowska M, Rose-John S, Mackiewicz A. Whole Cell Therapeutic Vaccine Modified With Hyper-IL6 for Combinational Treatment of Nonresected Advanced Melanoma. Medicine (Baltimore) 2015; 94:e853. [PMID: 26020391 PMCID: PMC4616404 DOI: 10.1097/md.0000000000000853] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
UNLABELLED Active specific immunotherapy of cancer requires an efficient induction and effector phase. The induction covers potent activation of anti-tumor response, whereas effector breaks the immunosuppression. We report efficacy of therapeutic melanoma vaccine (AGI-101H) used alone in advanced disease as a candidate for further combined treatment. In adjuvant setting in patients with resected metastases AGI-101H combined with surgery of recurring disease demonstrated long-term survival. Seventy-seven patients with nonresectable melanoma (8% IIIB, 21% IIIC, 71% IV) were enrolled. AGI-101H was administered 8× every 2 weeks, and then every month. At progression, maintenance was continued or induction was repeated and followed by maintenance. Median follow-up was 139.3 months. The median overall survival (OS) was 17.3 months; in patients with WHO 0-1 was 20.3 months. Complete response (CR) and partial response (PR) were observed in 19.4% and 9% of pts. Disease control rate was 54.5% of pts. The median CR+PR duration was 32 months. Reinduction was performed in 36.3% patients following disease progression with 46.6% of CR+PR. No grade 3/4 adverse events were observed. Treatment with AGI-101H of melanoma patients is safe and effective. AGI-101H is a good candidate for combinatorial treatment with immune check-points inhibitors or tumor hypoxia normalizators. TRIAL REGISTRATION EudraCT Number 2008-003373-40.
Collapse
Affiliation(s)
- Jacek Mackiewicz
- From the Chair of Medical Biotechnology, University of Medical Sciences (JM, ML, MK, MW, MZ, AM); Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre (JM, A-KD, MW, AM); Department of Medical and Experimental Oncology, Clinical Hospital of Poznan University of Medical Sciences, Poznan, Poland (JM); Interuniversity Institute for Biostatistics and Statistical Bioinformatics, Hasselt University, Diepenbeek, Belgium (TB); Biochemisches Institut, University Kiel, Kiel, Germany (SR-J); and BioContract Sp z o.o., Poznan, Poland (ML, MK, AM)
| | | | | | | | | | | | | | | | | |
Collapse
|
4252
|
Sullivan RJ, Flaherty KT. Pembrolizumab for Treatment of Patients with Advanced or Unresectable Melanoma. Clin Cancer Res 2015; 21:2892-7. [DOI: 10.1158/1078-0432.ccr-14-3061] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 03/06/2015] [Indexed: 11/16/2022]
|
4253
|
Birkeland AC, Brenner JC. Personalizing Medicine in Head and Neck Squamous Cell Carcinoma: The Rationale for Combination Therapies. ACTA ACUST UNITED AC 2015; 3. [PMID: 26913293 DOI: 10.18103/mra.v0i3.77] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Personalized medicine, in which individual tumor genetics drive the selection of targeted therapies and treatment plans for each patient, has recently emerged as the next generation of cancer therapy. Unfortunately, personalized medicine trials have had limited success in tumors that have complex combinations of disruptive genomic events, which drive differential responses to targeted therapies. Here, we will use head and neck squamous cell carcinoma as a model for genetically complex disease and discuss novel approaches to enhance personalized medicine trials for these complicated cases.
Collapse
Affiliation(s)
- Andrew C Birkeland
- Department of Otolaryngology - Head and Neck Surgery, The University of Michigan Medical School, Ann Arbor, MI
| | - J Chad Brenner
- Department of Otolaryngology - Head and Neck Surgery, The University of Michigan Medical School, Ann Arbor, MI; Comprehensive Cancer Center, The University of Michigan Medical School, Ann Arbor, MI
| |
Collapse
|
4254
|
Martin-Liberal J, Kordbacheh T, Larkin J. Safety of pembrolizumab for the treatment of melanoma. Expert Opin Drug Saf 2015; 14:957-64. [DOI: 10.1517/14740338.2015.1021774] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
4255
|
Abstract
Immune checkpoint therapy, which targets regulatory pathways in T cells to enhance antitumor immune responses, has led to important clinical advances and provided a new weapon against cancer. This therapy has elicited durable clinical responses and, in a fraction of patients, long-term remissions where patients exhibit no clinical signs of cancer for many years. The way forward for this class of novel agents lies in our ability to understand human immune responses in the tumor microenvironment. This will provide valuable information regarding the dynamic nature of the immune response and regulation of additional pathways that will need to be targeted through combination therapies to provide survival benefit for greater numbers of patients.
Collapse
Affiliation(s)
- Padmanee Sharma
- Department of Immunology, M.D. Anderson Cancer Center, Houston, TX, USA. Genitourinary Medical Oncology, M.D. Anderson Cancer Center, Houston, TX, USA.
| | - James P Allison
- Department of Immunology, M.D. Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
4256
|
Queirolo P, Picasso V, Spagnolo F. Combined BRAF and MEK inhibition for the treatment of BRAF-mutated metastatic melanoma. Cancer Treat Rev 2015; 41:519-26. [PMID: 25944484 DOI: 10.1016/j.ctrv.2015.04.010] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Revised: 04/20/2015] [Accepted: 04/22/2015] [Indexed: 02/07/2023]
Abstract
Combined BRAF and MEK inhibition out-performed BRAF inhibitor monotherapy in 3 randomized Phase 3 studies for BRAF-mutated metastatic melanoma patients and the combination of BRAF inhibitor dabrafenib with MEK inhibitor trametinib is now an FDA-approved treatment in this setting. Nevertheless, the majority of patients face progressive disease even when treated with the combination. Mechanisms of resistance to BRAF inhibition have been extensively investigated, whilst less is known about the specific mechanisms of resistance to combined therapy. The aim of this paper is to review the efficacy and safety of the combination of BRAF plus MEK inhibitors compared with BRAF inhibitor monotherapy and immunotherapy, as well as to discuss the existing evidence for the mechanisms of resistance to combined therapy and assess future treatment strategies to improve outcome based on data provided by clinical and translational research studies.
Collapse
Affiliation(s)
- Paola Queirolo
- Department of Medical Oncology, IRCCS San Martino, IST Istituto Nazionale per la Ricerca sul Cancro, Genova, Italy
| | - Virginia Picasso
- Department of Medical Oncology, IRCCS San Martino, IST Istituto Nazionale per la Ricerca sul Cancro, Genova, Italy
| | - Francesco Spagnolo
- Department of Plastic and Reconstructive Surgery, IRCCS San Martino, IST Istituto Nazionale per la Ricerca sul Cancro, Genova, Italy.
| |
Collapse
|
4257
|
Conry RM, Sullivan JC, Nabors LB. Ipilimumab-induced encephalopathy with a reversible splenial lesion. Cancer Immunol Res 2015; 3:598-601. [PMID: 25922203 DOI: 10.1158/2326-6066.cir-15-0035] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 03/18/2015] [Indexed: 11/16/2022]
Abstract
Ipilimumab, an anticytotoxic T-lymphocyte antigen (CTLA)-4 monoclonal antibody, is a first-line therapy for stage IV melanoma. Although high-grade immune-related adverse events occur in 25% of patients receiving ipilimumab, serious neurologic toxicity, primarily consisting of transient sensory and motor neuropathies, affects less than 1% of patients. We present a case report of a patient with melanoma who received high-dose ipilimumab at 10 mg/kg as first-line therapy for metastatic disease. After the third dose, the patient developed "mild" encephalopathy with a reversible splenial lesion (MERS) of the corpus callosum by MRI and neurogenic bladder, two novel immune-related adverse events during checkpoint inhibition. In addition to headache, delirium, and altered consciousness commonly seen with MERS, the patient also developed tremor, gait instability, paresthesias, and neurogenic bladder. The latter two symptoms were thought to represent sensory and autonomic neuropathies, respectively. The syndrome gradually resolved following intravenous methylprednisolone at 2 mg/kg divided twice daily for 5 days and a slow taper of oral prednisone over 8 weeks.
Collapse
Affiliation(s)
- Robert M Conry
- Division of Hematology Oncology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama.
| | - Joseph C Sullivan
- Neuroradiology Section, Department of Radiology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Louis B Nabors
- Neuro-Oncology Program, Department of Neurology, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
4258
|
Bezu L, Gomes-de-Silva LC, Dewitte H, Breckpot K, Fucikova J, Spisek R, Galluzzi L, Kepp O, Kroemer G. Combinatorial strategies for the induction of immunogenic cell death. Front Immunol 2015; 6:187. [PMID: 25964783 PMCID: PMC4408862 DOI: 10.3389/fimmu.2015.00187] [Citation(s) in RCA: 193] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Accepted: 04/06/2015] [Indexed: 12/12/2022] Open
Abstract
The term "immunogenic cell death" (ICD) is commonly employed to indicate a peculiar instance of regulated cell death (RCD) that engages the adaptive arm of the immune system. The inoculation of cancer cells undergoing ICD into immunocompetent animals elicits a specific immune response associated with the establishment of immunological memory. Only a few agents are intrinsically endowed with the ability to trigger ICD. These include a few chemotherapeutics that are routinely employed in the clinic, like doxorubicin, mitoxantrone, oxaliplatin, and cyclophosphamide, as well as some agents that have not yet been approved for use in humans. Accumulating clinical data indicate that the activation of adaptive immune responses against dying cancer cells is associated with improved disease outcome in patients affected by various neoplasms. Thus, novel therapeutic regimens that trigger ICD are urgently awaited. Here, we discuss current combinatorial approaches to convert otherwise non-immunogenic instances of RCD into bona fide ICD.
Collapse
Affiliation(s)
- Lucillia Bezu
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers , Paris , France ; U1138, INSERM , Paris , France ; Metabolomics and Cell Biology Platforms, Gustave Roussy Campus Cancer , Villejuif , France ; Faculté de Medecine, Université Paris-Sud , Le Kremlin-Bicêtre , France
| | - Ligia C Gomes-de-Silva
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers , Paris , France ; U1138, INSERM , Paris , France ; Metabolomics and Cell Biology Platforms, Gustave Roussy Campus Cancer , Villejuif , France ; Department of Chemistry, University of Coimbra , Coimbra , Portugal
| | - Heleen Dewitte
- Laboratory for General Biochemistry and Physical Pharmacy, Faculty of Pharmacy, Ghent University , Ghent , Belgium ; Laboratory of Molecular and Cellular Therapy, Vrije Universiteit Brussel , Jette , Belgium
| | - Karine Breckpot
- Laboratory of Molecular and Cellular Therapy, Vrije Universiteit Brussel , Jette , Belgium
| | - Jitka Fucikova
- Sotio a.c. , Prague , Czech Republic ; Department of Immunology, 2nd Faculty of Medicine, University Hospital Motol, Charles University , Prague , Czech Republic
| | - Radek Spisek
- Sotio a.c. , Prague , Czech Republic ; Department of Immunology, 2nd Faculty of Medicine, University Hospital Motol, Charles University , Prague , Czech Republic
| | - Lorenzo Galluzzi
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers , Paris , France ; U1138, INSERM , Paris , France ; Gustave Roussy Campus Cancer , Villejuif , France ; Université Paris Descartes , Paris , France ; Université Pierre et Marie Curie , Paris , France
| | - Oliver Kepp
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers , Paris , France ; U1138, INSERM , Paris , France ; Metabolomics and Cell Biology Platforms, Gustave Roussy Campus Cancer , Villejuif , France
| | - Guido Kroemer
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers , Paris , France ; U1138, INSERM , Paris , France ; Metabolomics and Cell Biology Platforms, Gustave Roussy Campus Cancer , Villejuif , France ; Department of Immunology, 2nd Faculty of Medicine, University Hospital Motol, Charles University , Prague , Czech Republic ; Université Paris Descartes , Paris , France ; Université Pierre et Marie Curie , Paris , France ; Pôle de Biologie, Hopitâl Européen George Pompidou, AP-HP , Paris , France
| |
Collapse
|
4259
|
Abstract
Melanoma is a common cancer in the Western world with an increasing incidence. Sun exposure is still considered to be the major risk factor for melanoma. The prognosis of patients with malignant (advanced-stage) melanoma differs widely between countries, but public campaigns advocating early detection have led to significant reductions in mortality rates. As well as sun exposure, distinct genetic alterations have been identified as associated with melanoma. For example, families with melanoma who have germline mutations in CDKN2A are well known, whereas the vast majority of sporadic melanomas have mutations in the mitogen-activated protein kinase cascade, which is the pathway with the highest oncogenic and therapeutic relevance for this disease. BRAF and NRAS mutations are typically found in cutaneous melanomas, whereas KIT mutations are predominantly observed in mucosal and acral melanomas. GNAQ and GNA11 mutations prevail in uveal melanomas. Additionally, the PI3K-AKT-PTEN pathway and the immune checkpoint pathways are important. The finding that programmed cell death protein 1 ligand 1 (PDL1) and PDL2 are expressed by melanoma cells, T cells, B cells and natural killer cells led to the recent development of programmed cell death protein 1 (PD1)-specific antibodies (for example, nivolumab and pembrolizumab). Alongside other new drugs - namely, BRAF inhibitors (vemurafenib and dabrafenib) and MEK inhibitors (trametinib and cobimetinib) - these agents are very promising and have been shown to significantly improve prognosis for patients with advanced-stage metastatic disease. Early signs are apparent that these new treatment modalities are also improving long-term clinical benefit and the quality of life of patients. This Primer summarizes the current understanding of melanoma, from mechanistic insights to clinical progress. For an illustrated summary of this Primer, visit: http://go.nature.com/vX2N9s.
Collapse
|
4260
|
Gangadhar TC, Salama AK. Clinical applications of PD-1-based therapy: a focus on pembrolizumab (MK-3475) in the management of melanoma and other tumor types. Onco Targets Ther 2015; 8:929-37. [PMID: 25960664 PMCID: PMC4410900 DOI: 10.2147/ott.s53164] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Preclinical work has led to an increased understanding of the immunomodulatory mechanisms involved in the regulation of the antitumor response in a variety of tumor types. PD-1 (programmed death 1) appears to be a key checkpoint involved in immune suppression in the tumor microenvironment, even in diseases not previously thought to be sensitive to immune manipulation. More recently, the subsequent clinical development of PD-1-based therapy has resulted in a major breakthrough in the field of oncology. Pembrolizumab, a humanized highly selective IgG4 anti-PD-1 monoclonal antibody, was recently approved for the treatment of advanced melanoma based on promising early-phase clinical data. Encouraging results have also been seen in other malignancies, and PD-1-targeted therapies are likely to markedly change the treatment landscape. Future work will center on rationally designed combination strategies in order to potentiate the antitumor immune response and overcome mechanisms of resistance.
Collapse
Affiliation(s)
- Tara C Gangadhar
- Division of Hematology/Oncology, Department of Medicine, Abramson Cancer Center of the University of Pennsylvania, Philadelphia, PA, USA
| | - April Ks Salama
- Division of Medical Oncology, Department of Medicine, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
4261
|
Läubli H, Balmelli C, Bossard M, Pfister O, Glatz K, Zippelius A. Acute heart failure due to autoimmune myocarditis under pembrolizumab treatment for metastatic melanoma. J Immunother Cancer 2015; 3:11. [PMID: 25901283 PMCID: PMC4404586 DOI: 10.1186/s40425-015-0057-1] [Citation(s) in RCA: 249] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 03/11/2015] [Indexed: 02/08/2023] Open
Abstract
Antibodies that stimulate the immune system by targeting inhibitory T cell receptors were successfully introduced into oncological practice and are capable to overcome tumor-induced immune evasion. In particular, targeting of the inhibitory receptors CTLA-4 and PD-1 or its ligand PD-L1 have been shown to be beneficial for patients with melanoma, renal cell cancer, non-small cell lung cancer and a growing list of other cancers with impressive response rates. Here, we report a severe, potentially life-threatening side effect of anti-PD-1 immunotherapy with pembrolizumab, which has not been previously described in the literature. A 73-year-old woman with metastatic uveal melanoma treated with pembrolizumab in third line developed severe heart failure due to pembrolizumab-mediated autoimmune myocarditis. Echocardiographic studies revealed a severely impaired left ventricular function with dyssynchrony. All tests for cardiotropic viruses were negative and histological analysis of a myocardial biopsy showed lymphocytic infiltration with a predominance of CD8 positive cells and a reduction of FOXP3 positive regulatory T cells. After initiation of corticosteroids and guideline-conform heart failure therapy, the symptoms rapidly improved and the left ventricular function recovered. While autoimmune myocarditis is a documented side effect of other checkpoint inhibitors, as for example ipilimumab and in one case with anti-PD-L1 antibody, it is not described for anti-PD-1-antibodies like pembrolizumab or nivolumab. As the FDA recently approved both pembrolizumab and nivolumab for melanoma progressing after anti-CTLA-4 treatment with ipilimumab, more patients will soon receive anti-PD-1 therapy. Thus, it is important to be aware of such rare, but severe immune-related adverse events.
Collapse
Affiliation(s)
- Heinz Läubli
- Division of Medical Oncology, University Hospital Basel, Basel, Switzerland.,Department of Biomedicine, Cancer Immunology Laboratory, University of Basel, Basel, Switzerland
| | - Cathrin Balmelli
- Division of Medical Oncology, University Hospital Basel, Basel, Switzerland
| | - Matthias Bossard
- Division of Cardiology, University Hospital Basel, Basel, Switzerland
| | - Otmar Pfister
- Division of Cardiology, University Hospital Basel, Basel, Switzerland
| | - Kathrin Glatz
- Institute of Pathology, University Hospital Basel, Basel, Switzerland
| | - Alfred Zippelius
- Division of Medical Oncology, University Hospital Basel, Basel, Switzerland.,Department of Biomedicine, Cancer Immunology Laboratory, University of Basel, Basel, Switzerland
| |
Collapse
|
4262
|
Emerging strategies to effectively target autophagy in cancer. Oncogene 2015; 35:1-11. [PMID: 25893285 DOI: 10.1038/onc.2015.99] [Citation(s) in RCA: 199] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Revised: 02/18/2015] [Accepted: 02/18/2015] [Indexed: 02/07/2023]
Abstract
Autophagy serves a dichotomous role in cancer and recent advances have helped delineate the appropriate settings where inhibiting or promoting autophagy may confer therapeutic efficacy in patients. Our evolving understanding of the molecular machinery responsible for the tightly controlled regulation of this homeostatic mechanism has begun to bear fruit in the way of autophagy-oriented clinical trials and promising lead compounds to modulate autophagy for therapeutic benefit. In this manuscript we review the recent preclinical and clinical therapeutic strategies that involve autophagy modulation in cancer.
Collapse
|
4263
|
Printz C. New immunotherapies show promise in some patients: physicians try to balance optimism with realistic expectations. Cancer 2015; 121:1155-6. [PMID: 25855330 DOI: 10.1002/cncr.28992] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
4264
|
Topalian SL, Drake CG, Pardoll DM. Immune checkpoint blockade: a common denominator approach to cancer therapy. Cancer Cell 2015; 27:450-61. [PMID: 25858804 PMCID: PMC4400238 DOI: 10.1016/j.ccell.2015.03.001] [Citation(s) in RCA: 3171] [Impact Index Per Article: 317.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Revised: 02/17/2015] [Accepted: 03/03/2015] [Indexed: 02/08/2023]
Abstract
The immune system recognizes and is poised to eliminate cancer but is held in check by inhibitory receptors and ligands. These immune checkpoint pathways, which normally maintain self-tolerance and limit collateral tissue damage during anti-microbial immune responses, can be co-opted by cancer to evade immune destruction. Drugs interrupting immune checkpoints, such as anti-CTLA-4, anti-PD-1, anti-PD-L1, and others in early development, can unleash anti-tumor immunity and mediate durable cancer regressions. The complex biology of immune checkpoint pathways still contains many mysteries, and the full activity spectrum of checkpoint-blocking drugs, used alone or in combination, is currently the subject of intense study.
Collapse
Affiliation(s)
- Suzanne L Topalian
- Department of Surgery, Sidney Kimmel Comprehensive Cancer Center and Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| | - Charles G Drake
- The Brady Urological Institute, Sidney Kimmel Comprehensive Cancer Center and Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Department of Oncology, Sidney Kimmel Comprehensive Cancer Center and Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Drew M Pardoll
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center and Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
4265
|
Sharma P, Allison JP. Immune checkpoint targeting in cancer therapy: toward combination strategies with curative potential. Cell 2015. [PMID: 25860605 DOI: 10.1016/j.cell.2015.03.030.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/01/2022]
Abstract
Research in two fronts has enabled the development of therapies that provide significant benefit to cancer patients. One area stems from a detailed knowledge of mutations that activate or inactivate signaling pathways that drive cancer development. This work triggered the development of targeted therapies that lead to clinical responses in the majority of patients bearing the targeted mutation, although responses are often of limited duration. In the second front are the advances in molecular immunology that unveiled the complexity of the mechanisms regulating cellular immune responses. These developments led to the successful targeting of immune checkpoints to unleash anti-tumor T cell responses, resulting in durable long-lasting responses but only in a fraction of patients. In this Review, we discuss the evolution of research in these two areas and propose that intercrossing them and increasing funding to guide research of combination of agents represent a path forward for the development of curative therapies for the majority of cancer patients.
Collapse
Affiliation(s)
- Padmanee Sharma
- Department of Immunology, MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Genitourinary Medical Oncology, MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - James P Allison
- Department of Immunology, MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
4266
|
Sharma P, Allison JP. Immune checkpoint targeting in cancer therapy: toward combination strategies with curative potential. Cell 2015; 161:205-14. [PMID: 25860605 PMCID: PMC5905674 DOI: 10.1016/j.cell.2015.03.030] [Citation(s) in RCA: 1769] [Impact Index Per Article: 176.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Indexed: 12/16/2022]
Abstract
Research in two fronts has enabled the development of therapies that provide significant benefit to cancer patients. One area stems from a detailed knowledge of mutations that activate or inactivate signaling pathways that drive cancer development. This work triggered the development of targeted therapies that lead to clinical responses in the majority of patients bearing the targeted mutation, although responses are often of limited duration. In the second front are the advances in molecular immunology that unveiled the complexity of the mechanisms regulating cellular immune responses. These developments led to the successful targeting of immune checkpoints to unleash anti-tumor T cell responses, resulting in durable long-lasting responses but only in a fraction of patients. In this Review, we discuss the evolution of research in these two areas and propose that intercrossing them and increasing funding to guide research of combination of agents represent a path forward for the development of curative therapies for the majority of cancer patients.
Collapse
Affiliation(s)
- Padmanee Sharma
- Department of Immunology, MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Genitourinary Medical Oncology, MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - James P Allison
- Department of Immunology, MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
4267
|
Amaria RN, Reuben A, Cooper ZA, Wargo JA. Update on use of aldesleukin for treatment of high-risk metastatic melanoma. Immunotargets Ther 2015; 4:79-89. [PMID: 27471714 PMCID: PMC4918260 DOI: 10.2147/itt.s61590] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
High-dose interleukin-2 has been used for the treatment of metastatic melanoma since 1998 based on data proving durable complete responses in up to 10% of treated patients. The immunomodulatory effects of this critical cytokine have been instrumental in the development of immunotherapy for melanoma and other cancers. However, with the advent of new therapies, its use as a front-line agent has come into question. Nonetheless, there is still a role for interleukin-2 as monotherapy, as well as in combination with other agents and in clinical trials. In this article, we review preclinical and clinical data regarding interleukin-2, its pharmacology and mechanism of action, its toxicity profile, and its use in ongoing and planned clinical trials. We also explore the future of this agent within the treatment landscape for melanoma.
Collapse
Affiliation(s)
- Rodabe N Amaria
- Department of Melanoma Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Alexandre Reuben
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Zachary A Cooper
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jennifer A Wargo
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
4268
|
Hughes J, Vudattu N, Sznol M, Gettinger S, Kluger H, Lupsa B, Herold KC. Precipitation of autoimmune diabetes with anti-PD-1 immunotherapy. Diabetes Care 2015; 38:e55-7. [PMID: 25805871 PMCID: PMC4370325 DOI: 10.2337/dc14-2349] [Citation(s) in RCA: 237] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- Jing Hughes
- Department of Immunobiology, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT Section of Endocrinology and Metabolism, Yale University School of Medicine, New Haven, CT
| | - Nalini Vudattu
- Department of Immunobiology, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT
| | - Mario Sznol
- Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT
| | - Scott Gettinger
- Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT
| | - Harriet Kluger
- Department of Internal Medicine, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT
| | - Beatrice Lupsa
- Section of Endocrinology and Metabolism, Yale University School of Medicine, New Haven, CT
| | - Kevan C Herold
- Department of Immunobiology, Section of Medical Oncology, Yale University School of Medicine, New Haven, CT Section of Endocrinology and Metabolism, Yale University School of Medicine, New Haven, CT
| |
Collapse
|
4269
|
Buqué A, Bloy N, Aranda F, Castoldi F, Eggermont A, Cremer I, Fridman WH, Fucikova J, Galon J, Marabelle A, Spisek R, Tartour E, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch: Immunomodulatory monoclonal antibodies for oncological indications. Oncoimmunology 2015; 4:e1008814. [PMID: 26137403 PMCID: PMC4485728 DOI: 10.1080/2162402x.2015.1008814] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2015] [Accepted: 01/12/2015] [Indexed: 12/14/2022] Open
Abstract
Immunomodulatory monoclonal antibodies (mAbs) differ from their tumor-targeting counterparts because they exert therapeutic effects by directly interacting with soluble or (most often) cellular components of the immune system. Besides holding promise for the treatment of autoimmune and inflammatory disorders, immunomodulatory mAbs have recently been shown to constitute a potent therapeutic weapon against neoplastic conditions. One class of immunomodulatory mAbs operates by inhibiting safeguard systems that are frequently harnessed by cancer cells to establish immunological tolerance, the so-called "immune checkpoints." No less than 3 checkpoint-blocking mAbs have been approved worldwide for use in oncological indications, 2 of which during the past 12 months. These molecules not only mediate single-agent clinical activity in patients affected by specific neoplasms, but also significantly boost the efficacy of several anticancer chemo-, radio- or immunotherapies. Here, we summarize recent advances in the development of checkpoint-blocking mAbs, as well as of immunomodulatory mAbs with distinct mechanisms of action.
Collapse
Key Words
- CRC, colorectal carcinoma
- CTLA4, cytotoxic T lymphocyte-associated protein 4
- FDA, Food and Drug Administration
- IL, interleukin
- KIR, killer cell immunoglobulin-like receptor
- MEDI4736
- MPDL3280A
- NK, natural killer
- NSCLC, non-small cell lung carcinoma
- PD-1, programmed cell death 1
- RCC, renal cell carcinoma
- TGFβ1, transforming growth factor β1
- TLR, Toll-like receptor
- TNFRSF, tumor necrosis factor receptor superfamily
- Treg, regulatory T cell
- ipilimumab
- mAb, monoclonal antibody
- nivolumab
- pembrolizumab
- urelumab
Collapse
Affiliation(s)
- Aitziber Buqué
- Gustave Roussy Cancer Campus; Villejuif, France
- INSERM, U1138; Paris, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Center de Recherche des Cordeliers; Paris, France
| | - Norma Bloy
- Gustave Roussy Cancer Campus; Villejuif, France
- INSERM, U1138; Paris, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Center de Recherche des Cordeliers; Paris, France
- Faculté de Medicine, Université Paris Sud/Paris XI; Le Kremlin-Bicêtre, France
| | - Fernando Aranda
- Group of Immune receptors of the Innate and Adaptive System, Institut d'Investigacions Biomédiques August Pi i Sunyer (IDIBAPS); Barcelona, Spain
| | - Francesca Castoldi
- Gustave Roussy Cancer Campus; Villejuif, France
- INSERM, U1138; Paris, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Center de Recherche des Cordeliers; Paris, France
- Faculté de Medicine, Université Paris Sud/Paris XI; Le Kremlin-Bicêtre, France
- Sotio a.c.; Prague, Czech Republic
| | | | - Isabelle Cremer
- INSERM, U1138; Paris, France
- Equipe 13, Center de Recherche des Cordeliers; Paris, France
- Université Pierre et Marie Curie/Paris VI; Paris, France
| | - Wolf Hervé Fridman
- INSERM, U1138; Paris, France
- Université Pierre et Marie Curie/Paris VI; Paris, France
- Dept. of Immunology, 2nd Faculty of Medicine and University Hospital Motol, Charles University; Prague, Czech Republic
| | - Jitka Fucikova
- Sotio a.c.; Prague, Czech Republic
- Dept. of Immunology, 2nd Faculty of Medicine and University Hospital Motol, Charles University; Prague, Czech Republic
| | - Jérôme Galon
- INSERM, U1138; Paris, France
- Université Pierre et Marie Curie/Paris VI; Paris, France
- Laboratory of Integrative Cancer Immunology, Center de Recherche des Cordeliers; Paris, France
- Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France
| | - Aurélien Marabelle
- Gustave Roussy Cancer Campus; Villejuif, France
- INSERM, U1015, CICBT507; Villejuif, France
| | - Radek Spisek
- Sotio a.c.; Prague, Czech Republic
- Equipe 13, Center de Recherche des Cordeliers; Paris, France
| | - Eric Tartour
- Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France
- INSERM, U970; Paris, France
- Paris-Cardiovascular Research Center (PARCC); Paris, France
- Service d'Immunologie Biologique, Hôpital Européen Georges Pompidou (HEGP); AP-HP; Paris, France
| | - Laurence Zitvogel
- Gustave Roussy Cancer Campus; Villejuif, France
- INSERM, U1015, CICBT507; Villejuif, France
| | - Guido Kroemer
- INSERM, U1138; Paris, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Center de Recherche des Cordeliers; Paris, France
- Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France
- Pôle de Biologie, Hôpital Européen Georges Pompidou; AP-HP; Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus; Villejuif, France
| | - Lorenzo Galluzzi
- Gustave Roussy Cancer Campus; Villejuif, France
- INSERM, U1138; Paris, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Center de Recherche des Cordeliers; Paris, France
- Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France
| |
Collapse
|
4270
|
Doma V, Gulya E. [Genetic diversity and immunological characteristics of malignant melanoma: the therapeutic spectrum]. Orv Hetil 2015; 156:583-91. [PMID: 25845317 DOI: 10.1556/oh.2015.30122] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Malignant melanoma, originating from pigment cells, is a highly aggressive tumour affecting patients of any age group. Its incidence is rapidly growing. The most common form can be easily diagnosed by any physician. There are some well-known genetic (skin-, eye-, hair colour, naevi, melanoma in the personal/family history) and environmental (ultraviolet radiation) predisposing factors. Treatment is based on early diagnosis and excision. When metastasis occurs, the traditional chemo- and radiotherapy gives a low response rate. Recently some newly approved targeted therapies and immunomodulant drugs have become available. This review focuses on the classification and novel therapeutic approaches of malignant melanoma to provide guidance to clinicians.
Collapse
Affiliation(s)
- Viktória Doma
- Semmelweis Egyetem, Általános Orvostudományi Kar II. Patológiai Intézet Budapest Fejér Megyei Szent György Egyetemi Oktató Kórház Bőrgyógyászati Osztály Székesfehérvár
| | - Ernő Gulya
- Magyar Honvédség Egészségügyi Központ Honvédkórház Budapest
| |
Collapse
|
4271
|
|
4272
|
Abstract
It has long been understood that the immune system has intrinsic anti-tumour activity in humans, and that a key mechanism of tumour progression is the ability of a tumour to escape this immune surveillance. A number of attempts have been made to harness this anti-tumour immunity in both solid tumour oncology and haematological malignancies with variable success. Examples include the use of allogeneic stem cell transplantation and donor lymphocyte infusion in haematological cancer and vaccine studies in solid tumours. Enhanced signalling of the Programmed cell death-1 (PDCD1, PD-1)/cytotoxic T-lymphocyte-associated protein 4 (CTLA4) 'immune checkpoint' pathway has emerged recently as a critical mechanism by which tumours can escape the natural anti-tumour immune response. As such, novel therapies have been developed to help enhance this natural immunity by switching off the PDCD1/CTLA4 immune checkpoint pathway. The following review will discuss the pathobiology of these pathways and the exciting new data now available in lymphoid malignancies.
Collapse
Affiliation(s)
- Toby A Eyre
- Department of Haematology, Oxford University Hospitals NHS Trust, Oxford, UK
| | - Graham P Collins
- Department of Haematology, Oxford University Hospitals NHS Trust, Oxford, UK
| |
Collapse
|
4273
|
Mahoney KM, Freeman GJ, McDermott DF. The Next Immune-Checkpoint Inhibitors: PD-1/PD-L1 Blockade in Melanoma. Clin Ther 2015; 37:764-82. [PMID: 25823918 DOI: 10.1016/j.clinthera.2015.02.018] [Citation(s) in RCA: 443] [Impact Index Per Article: 44.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2014] [Revised: 02/15/2015] [Accepted: 02/17/2015] [Indexed: 12/31/2022]
Abstract
PURPOSE Blocking the interaction between the programmed cell death (PD)-1 protein and one of its ligands, PD-L1, has been reported to have impressive antitumor responses. Therapeutics targeting this pathway are currently in clinical trials. Pembrolizumab and nivolumab are the first of this anti-PD-1 pathway family of checkpoint inhibitors to gain accelerated approval from the US Food and Drug Administration (FDA) for the treatment of ipilimumab-refractory melanoma. Nivolumab has been associated with improved overall survival compared with dacarbazine in patients with previously untreated wild-type serine/threonine-protein kinase B-raf proto-oncogene BRAF melanoma. Although the most mature data are in the treatment of melanoma, the FDA has granted approval of nivolumab for squamous cell lung cancer and the breakthrough therapy designation to immune- checkpoint inhibitors for use in other cancers: nivolumab, an anti-PD-1 monoclonal antibody, for Hodgkin lymphoma, and MPDL-3280A, an anti-PD-L1 monoclonal antibody, for bladder cancer and non-small cell lung cancer. Here we review the literature on PD-1 and PD-L1 blockade and focus on the reported clinical studies that have included patients with melanoma. METHODS PubMed was searched to identify relevant clinical studies of PD-1/PD-L1-targeted therapies in melanoma. A review of data from the current trials on clinicaltrial.gov was incorporated, as well as data presented in abstracts at the 2014 annual meeting of the American Society of Clinical Oncology, given the limited number of published clinical trials on this topic. FINDINGS The anti-PD-1 and anti-PD-L1 agents have been reported to have impressive antitumor effects in several malignancies, including melanoma. The greatest clinical activity in unselected patients has been seen in melanoma. Tumor expression of PD-L1 is a suggestive, but inadequate, biomarker predictive of response to immune-checkpoint blockade. However, tumors expressing little or no PD-L1 are less likely to respond to PD-1 pathway blockade. Combination checkpoint blockade with PD-1 plus cytotoxic T-lymphocyte antigen (CTLA)-4 blockade appears to improve response rates in patients who are less likely to respond to single-checkpoint blockade. Toxicity with PD-1 blocking agents is less than the toxicity with previous immunotherapies (eg, interleukin 2, CTLA-4 blockade). Certain adverse events can be severe and potentially life threatening, but most can be prevented or reversed with close monitoring and appropriate management. IMPLICATIONS This family of immune-checkpoint inhibitors benefits not only patients with metastatic melanoma but also those with historically less responsive tumor types. Although a subset of patients responds to single-agent blockade, the initial trial of checkpoint-inhibitor combinations has reported a potential to improve response rates. Combination therapies appear to be a means of increasing response rates, albeit with increased immune-related adverse events. As these treatments become available to patients, education regarding the recognition and management of immune-related effects of immune-checkpoint blockade will be essential for maximizing clinical benefit.
Collapse
Affiliation(s)
- Kathleen M Mahoney
- Division of Hematology and Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts; Department of Medical Oncology, Harvard Medical School, Dana Farber Cancer Institute, Boston, Massachusetts.
| | - Gordon J Freeman
- Department of Medical Oncology, Harvard Medical School, Dana Farber Cancer Institute, Boston, Massachusetts
| | - David F McDermott
- Division of Hematology and Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
4274
|
Flaherty KT. When are signal transduction targeted therapies acting as immunotherapy? Cancer Biol Ther 2015; 16:645-7. [PMID: 25807058 DOI: 10.1080/15384047.2015.1030553] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Affiliation(s)
- Keith T Flaherty
- a Massachusetts General Hospital Cancer Center ; Boston , MA , USA
| |
Collapse
|
4275
|
Madorsky Rowdo FP, Baron A, Urrutia M, Mordoh J. Immunotherapy in Cancer: A Combat between Tumors and the Immune System; You Win Some, You Lose Some. Front Immunol 2015; 6:127. [PMID: 25859247 PMCID: PMC4374472 DOI: 10.3389/fimmu.2015.00127] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 03/08/2015] [Indexed: 12/18/2022] Open
Abstract
Cancer immunotherapy has emerged as a treatment modality, mainly as the result of discoveries in the immune response regulation, including mechanisms that turn off immune responses. Immunogenic cutaneous melanoma is a canonical model for therapeutic immunotherapy studies. “Passive” immunotherapy with monoclonal antibodies (mAbs) has outpaced “active” immunotherapy with anti-tumor vaccines, and mAbs that antagonize the off responses have been recently introduced in clinical practice. Despite these recent successes, many unresolved practical and theoretical questions remain. Notably unknown are the identity of the lymphocytes that eliminate tumor cells, which white cells enter into tumors, through which endothelium, in what order, and how they perform their task. The parameters of size and location that could be used to determine in which tumors the immune response may be sufficient to eradicate the tumor are yet unknown. Immunotherapy has been so far more efficient to treat solid and hematologic tumors located outside the central nervous system, than primary brain tumors and brain metastases. In contrast to recent advances with mAbs, anti-tumor vaccine development has been lagging behind. The multiplicity of antigens that must be targeted to achieve significant clinical response is partially responsible for this lag, especially in melanoma, one of the most mutated tumors. Further hampering vaccination results is the fact that tumor elimination by the immune system is the result of a race between tumors with different growth rates and the relatively slow development of the adaptive immune response. The enhancement of the native arm of the immune response or the administration of targeted chemotherapy to slow tumor development, are approaches that should be studied. Finally, criteria used to analyze patient response to immunotherapeutic treatments must be perfected, and the patient populations that could benefit the most from this approach must be better defined.
Collapse
Affiliation(s)
| | - Antonela Baron
- Laboratorio de Cancerología, Fundación Instituto Leloir - IIBBA-CONICET , Buenos Aires , Argentina
| | - Mariela Urrutia
- Laboratorio de Cancerología, Fundación Instituto Leloir - IIBBA-CONICET , Buenos Aires , Argentina
| | - José Mordoh
- Laboratorio de Cancerología, Fundación Instituto Leloir - IIBBA-CONICET , Buenos Aires , Argentina ; Centro de Investigaciones Oncológicas, Fundación Cáncer and Instituto Alexander Fleming , Buenos Aires , Argentina
| |
Collapse
|
4276
|
Lesokhin AM, Callahan MK, Postow MA, Wolchok JD. On being less tolerant: Enhanced cancer immunosurveillance enabled by targeting checkpoints and agonists of T cell activation. Sci Transl Med 2015; 7:280sr1. [DOI: 10.1126/scitranslmed.3010274] [Citation(s) in RCA: 123] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
4277
|
Weber JS, D'Angelo SP, Minor D, Hodi FS, Gutzmer R, Neyns B, Hoeller C, Khushalani NI, Miller WH, Lao CD, Linette GP, Thomas L, Lorigan P, Grossmann KF, Hassel JC, Maio M, Sznol M, Ascierto PA, Mohr P, Chmielowski B, Bryce A, Svane IM, Grob JJ, Krackhardt AM, Horak C, Lambert A, Yang AS, Larkin J. Nivolumab versus chemotherapy in patients with advanced melanoma who progressed after anti-CTLA-4 treatment (CheckMate 037): a randomised, controlled, open-label, phase 3 trial. Lancet Oncol 2015; 16:375-84. [PMID: 25795410 DOI: 10.1016/s1470-2045(15)70076-8] [Citation(s) in RCA: 2089] [Impact Index Per Article: 208.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Nivolumab, a fully human IgG4 PD-1 immune checkpoint inhibitor antibody, can result in durable responses in patients with melanoma who have progressed after ipilimumab and BRAF inhibitors. We assessed the efficacy and safety of nivolumab compared with investigator's choice of chemotherapy (ICC) as a second-line or later-line treatment in patients with advanced melanoma. METHODS In this randomised, controlled, open-label, phase 3 trial, we recruited patients at 90 sites in 14 countries. Eligible patients were 18 years or older, had unresectable or metastatic melanoma, and progressed after ipilimumab, or ipilimumab and a BRAF inhibitor if they were BRAF(V 600) mutation-positive. Participating investigators randomly assigned (with an interactive voice response system) patients 2:1 to receive an intravenous infusion of nivolumab 3 mg/kg every 2 weeks or ICC (dacarbazine 1000 mg/m(2) every 3 weeks or paclitaxel 175 mg/m(2) combined with carboplatin area under the curve 6 every 3 weeks) until progression or unacceptable toxic effects. We stratified randomisation by BRAF mutation status, tumour expression of PD-L1, and previous best overall response to ipilimumab. We used permuted blocks (block size of six) within each stratum. Primary endpoints were the proportion of patients who had an objective response and overall survival. Treatment was given open-label, but those doing tumour assessments were masked to treatment assignment. We assessed objective responses per-protocol after 120 patients had been treated with nivolumab and had a minimum follow-up of 24 weeks, and safety in all patients who had had at least one dose of treatment. The trial is closed and this is the first interim analysis, reporting the objective response primary endpoint. This study is registered with ClinicalTrials.gov, number NCT01721746. FINDINGS Between Dec 21, 2012, and Jan 10, 2014, we screened 631 patients, randomly allocating 272 patients to nivolumab and 133 to ICC. Confirmed objective responses were reported in 38 (31·7%, 95% CI 23·5-40·8) of the first 120 patients in the nivolumab group versus five (10·6%, 3·5-23·1) of 47 patients in the ICC group. Grade 3-4 adverse events related to nivolumab included increased lipase (three [1%] of 268 patients), increased alanine aminotransferase, anaemia, and fatigue (two [1%] each); for ICC, these included neutropenia (14 [14%] of 102), thrombocytopenia (six [6%]), and anaemia (five [5%]). We noted grade 3-4 drug-related serious adverse events in 12 (5%) nivolumab-treated patients and nine (9%) patients in the ICC group. No treatment-related deaths occurred. INTERPRETATION Nivolumab led to a greater proportion of patients achieving an objective response and fewer toxic effects than with alternative available chemotherapy regimens for patients with advanced melanoma that has progressed after ipilimumab or ipilimumab and a BRAF inhibitor. Nivolumab represents a new treatment option with clinically meaningful durable objective responses in a population of high unmet need. FUNDING Bristol-Myers Squibb.
Collapse
Affiliation(s)
| | - Sandra P D'Angelo
- Memorial Sloan-Kettering Cancer Center and Weill Cornell Medical College, New York, NY, USA
| | - David Minor
- California Pacific Center for Melanoma Research, San Francisco, CA, USA
| | | | - Ralf Gutzmer
- Medizinische Hochschule Hannover, Hannover, Germany
| | - Bart Neyns
- Universitair Ziekenhuis Brussel, Brussels, Belgium
| | | | | | - Wilson H Miller
- Segal Cancer Centre, Jewish General Hospital, McGill University, Montreal, QC, Canada
| | | | | | - Luc Thomas
- Centre Hospitalier Universitaire de Lyon, Lyon, France
| | | | | | - Jessica C Hassel
- German Cancer Research Centre University Hospital, Heidelberg, Germany
| | - Michele Maio
- Medical Oncology and Immunotherapy, University Hospital of Siena, Istituto Toscano Tumori, Siena, Italy
| | | | | | - Peter Mohr
- Elbe Kliniken Buxtehude, Buxtehude, Germany
| | | | - Alan Bryce
- Department of Medicine, Mayo Clinic, Scottsdale, AZ, USA
| | - Inge M Svane
- Department of Oncology, Herlev Hospital, Copenhagen, Denmark
| | | | - Angela M Krackhardt
- Technische Universität München School of Medicine, II Medical Department, Munich, Germany
| | | | | | | | | |
Collapse
|
4278
|
Pauken KE, Wherry EJ. Overcoming T cell exhaustion in infection and cancer. Trends Immunol 2015; 36:265-76. [PMID: 25797516 DOI: 10.1016/j.it.2015.02.008] [Citation(s) in RCA: 839] [Impact Index Per Article: 83.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2015] [Revised: 02/24/2015] [Accepted: 02/24/2015] [Indexed: 12/18/2022]
Abstract
Inhibitors of the Programmed Cell Death 1: Programmed Cell Death 1 ligand 1 (PD-1:PD-L1) pathway, a central regulator of T cell exhaustion, have been recently shown to be effective for treatment of different cancers. However, clinical responses are mixed, highlighting the need to better understand the mechanisms of action of PD-1:PD-L1, the role of this pathway in immunity to different tumors, and the molecular and cellular effects of PD-1 blockade. Here, we review the molecular regulation of T cell exhaustion, placing recent findings on PD-1 blockade therapies in cancer in the context of the broader understanding of the roles of the PD-1:PD-L1 pathway in T cell exhaustion during chronic infection. We discuss the current understanding of the mechanisms involved in reversing T cell exhaustion, and outline critical areas of focus for future research, both basic and clinical.
Collapse
Affiliation(s)
- Kristen E Pauken
- Institute for Immunology and Department of Microbiology, University of Pennsylvania Perelman School of Medicine, 421 Curie Blvd, Philadelphia, PA 19104, USA
| | - E John Wherry
- Institute for Immunology and Department of Microbiology, University of Pennsylvania Perelman School of Medicine, 421 Curie Blvd, Philadelphia, PA 19104, USA.
| |
Collapse
|
4279
|
Abstract
Immunomodulatory nucleic acids have extraordinary promise for treating disease, yet clinical progress has been limited by a lack of tools to safely increase activity in patients. Immunomodulatory nucleic acids act by agonizing or antagonizing endosomal toll-like receptors (TLR3, TLR7/8, and TLR9), proteins involved in innate immune signaling. Immunomodulatory spherical nucleic acids (SNAs) that stimulate (immunostimulatory, IS-SNA) or regulate (immunoregulatory, IR-SNA) immunity by engaging TLRs have been designed, synthesized, and characterized. Compared with free oligonucleotides, IS-SNAs exhibit up to 80-fold increases in potency, 700-fold higher antibody titers, 400-fold higher cellular responses to a model antigen, and improved treatment of mice with lymphomas. IR-SNAs exhibit up to eightfold increases in potency and 30% greater reduction in fibrosis score in mice with nonalcoholic steatohepatitis (NASH). Given the clinical potential of SNAs due to their potency, defined chemical nature, and good tolerability, SNAs are attractive new modalities for developing immunotherapies.
Collapse
|
4280
|
The place of targeted agents in the treatment of elderly patients with metastatic colorectal cancer. Cancers (Basel) 2015; 7:439-49. [PMID: 25782007 PMCID: PMC4381267 DOI: 10.3390/cancers7010439] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 03/02/2015] [Accepted: 03/06/2015] [Indexed: 01/05/2023] Open
Abstract
Despite the high prevalence of colorectal cancer in a continuously aging population and the substantial advances in the treatment of metastatic disease during the past decade, the treatment of elderly patients with advanced, unresectable or metastatic colorectal cancer is a clearly unmet need. Since older patients are under-represented or even excluded from randomized trials, the evidence that oncologists use as guidance is weak. However, small prospective studies, pooled analyses and observational studies show that combination approaches are safe, efficacious and feasible in the geriatric population with metastatic colorectal cancer. The use of biologic agents targeting angiogenesis and the epidermal growth factor receptor, which have been shown to clearly improve outcomes in multiple prospective trials in patients with advanced colorectal cancer, is a vital component of the aforementioned combination approaches. Herein, we review all available data concerning the management of elderly patients with these agents and underscore the differences between this age subgroup and younger patients.
Collapse
|
4281
|
Abstract
The treatment and outcomes for people with metastatic melanoma have changed considerably in the past few years with the introduction of targeted anticancer drugs. About half of the patients with metastatic melanoma will have activating mutations in the BRAF gene. These people may benefit from a BRAF inhibitor (vemurafenib or dabrafenib) or a MEK inhibitor (trametinib). Addition of a MEK inhibitor to a BRAF inhibitor improves progression-free survival and alters the adverse effect profile. Ipilimumab is another drug indicated for metastatic melanoma. It works by altering the patient's own immune response to the tumour. Toxicities are common with these drugs and include arthralgias, fatigue, photosensitivity, squamous cell carcinomas, fever, diarrhoea, pruritus and immune-related adverse effects.
Collapse
|
4282
|
Phase II DeCOG-study of ipilimumab in pretreated and treatment-naïve patients with metastatic uveal melanoma. PLoS One 2015; 10:e0118564. [PMID: 25761109 PMCID: PMC4356548 DOI: 10.1371/journal.pone.0118564] [Citation(s) in RCA: 191] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Accepted: 01/16/2015] [Indexed: 12/30/2022] Open
Abstract
Purpose Up to 50% of patients with uveal melanoma (UM) develop metastatic disease with limited treatment options. The immunomodulating agent ipilimumab has shown an overall survival (OS) benefit in patients with cutaneous metastatic melanoma in two phase III trials. As patients with UM were excluded in these studies, the Dermatologic Cooperative Oncology Group (DeCOG) conducted a phase II to assess the efficacy and safety of ipilimumab in patients with metastatic UM. Patients and Methods We undertook a multicenter phase II study in patients with different subtypes of metastatic melanoma. Here we present data on patients with metastatic UM (pretreated and treatment-naïve) who received up to four cycles of ipilimumab administered at a dose of 3 mg/kg in 3 week intervals. Tumor assessments were conducted at baseline, weeks 12, 24, 36 and 48 according to RECIST 1.1 criteria. Adverse events (AEs), including immune-related AEs were graded according to National Cancer Institute Common Toxicity Criteria (CTC) v.4.0. Primary endpoint was the OS rate at 12 months. Results Forty five pretreated (85%) and eight treatment-naïve (15%) patients received at least one dose of ipilimumab. 1-year and 2-year OS rates were 22% and 7%, respectively. Median OS was 6.8 months (95% CI 3.7–8.1), median progression-free survival 2.8 months (95% CI 2.5–2.9). The disease control rate at weeks 12 and 24 was 47% and 21%, respectively. Sixteen patients had stable disease (47%), none experienced partial or complete response. Treatment-related AEs were observed in 35 patients (66%), including 19 grade 3–4 events (36%). One drug-related death due to pancytopenia was observed. Conclusions Ipilimumab has very limited clinical activity in patients with metastatic UM. Toxicity was manageable when treated as per protocol-specific guidelines. Trial Registration ClinicalTrials.gov NCT01355120
Collapse
|
4283
|
Johnson DB, Peng C, Sosman JA. Nivolumab in melanoma: latest evidence and clinical potential. Ther Adv Med Oncol 2015; 7:97-106. [PMID: 25755682 DOI: 10.1177/1758834014567469] [Citation(s) in RCA: 107] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Melanoma has historically been considered a refractory disease with few if any options in the advanced/metastatic setting. Advances in both immune and genetically targeted treatment approaches have revolutionized the spectrum of treatment options for melanoma patients over the last several years. Recently, checkpoint inhibition has become a major focus in the immune-based therapy of cancer, especially melanoma. This concept involves inhibition of regulatory cell surface molecules which act normally to dampen or modulate T-cell activation. Cancer, including melanoma, takes advantage of this physiologic mechanism to turn off T-cell activation and prevent effective T-cell antitumor responses. Checkpoint inhibitors such as anti cytotoxic T-lymphocyte antigen 4 (anti-CTLA-4) and anti programmed death-1 (PD-1) can reverse this immune suppression and release T-cell activation. Nivolumab, a monoclonal antibody to the PD-1 receptor, promotes antitumor immunity by removing this key negative regulator of T-cell activation. In phase I/II studies, promising activity and safety have been observed and ongoing phase III trials are comparing nivolumab with other standard of care therapies (chemotherapy, ipilimumab). Efficacy may be even further increased when used in combination with ipilimumab (albeit with increased toxicity). In contrast to typical short-lived responses with cancer therapy in metastatic solid tumors, many responses induced by nivolumab appear durable. In this review, we discuss the evolution of immune therapy in melanoma leading to the development of nivolumab, the clinical experience with this agent, and its future development and clinical potential.
Collapse
Affiliation(s)
- Douglas B Johnson
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, 777 Preston Research Building, 2220 Pierce Avenue, Nashville, TN 37232, USA
| | - Chengwei Peng
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jeffrey A Sosman
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
4284
|
Castillo JJ, Olszewski AJ, Hunter ZR, Kanan S, Meid K, Treon SP. Incidence of secondary malignancies among patients with Waldenström macroglobulinemia: An analysis of the SEER database. Cancer 2015; 121:2230-6. [DOI: 10.1002/cncr.29334] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2015] [Revised: 02/02/2015] [Accepted: 02/12/2015] [Indexed: 11/11/2022]
Affiliation(s)
- Jorge J. Castillo
- Bing Center for Waldenström Macroglobulinemia; Dana-Farber Cancer Institute; Boston Massachusetts
- Harvard Medical School; Boston Massachusetts
| | - Adam J. Olszewski
- Division of Hematology and Oncology; Memorial Hospital of Rhode Island, Warren Alpert Medical School of Brown University; Pawtucket Rhode Island
| | - Zachary R. Hunter
- Bing Center for Waldenström Macroglobulinemia; Dana-Farber Cancer Institute; Boston Massachusetts
| | - Sandra Kanan
- Bing Center for Waldenström Macroglobulinemia; Dana-Farber Cancer Institute; Boston Massachusetts
| | - Kirsten Meid
- Bing Center for Waldenström Macroglobulinemia; Dana-Farber Cancer Institute; Boston Massachusetts
| | - Steven P. Treon
- Bing Center for Waldenström Macroglobulinemia; Dana-Farber Cancer Institute; Boston Massachusetts
- Harvard Medical School; Boston Massachusetts
| |
Collapse
|
4285
|
Abstract
In patients with metastatic cancer, therapeutic anticancer vaccines are rarely associated with objective antitumor responses; so, many investigators have focused on progression-free survival (PFS) as a key endpoint for clinical trials. However, it is not clear that PFS is a surrogate for overall survival (OS), and OS may be a more appropriate endpoint because of the effects on long-term memory in the adaptive immune system. Recently, reported vaccine trials were reviewed to determine their primary and secondary endpoints and results. Randomized trials testing sipuleucel-T and prostvac-vf in prostate cancer and ipilimumab and eltrapuldencel-T in melanoma were associated with low objective response rates, no improvement in PFS, but statistically significant improvement in OS. Although compared with PFS, it takes longer to get a final result when OS is the primary endpoint; there is increasing evidence that if long-term memory recognition of tumor-associated antigens is the mechanism of action of an investigational product, then OS may be the only valid clinical endpoint for efficacy.
Collapse
|
4286
|
Curti BD, Urba WJ. Clinical deployment of antibodies for treatment of melanoma. Mol Immunol 2015; 67:18-27. [PMID: 25746916 DOI: 10.1016/j.molimm.2015.01.025] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Revised: 01/23/2015] [Accepted: 01/26/2015] [Indexed: 01/15/2023]
Abstract
The concept of using immunotherapy to treat melanoma has existed for decades. The rationale comes from the knowledge that many patients with melanoma have endogenous immune responses against their tumor cells and clinically meaningful tumor regression can be achieved in a minority of patients using cytokines such as interleukin-2 and adoptive cellular therapy. In the last 5 years there has been a revolution in the clinical management of melanoma in large measure based on the development of antibodies that influence T cell regulatory pathways by overcoming checkpoint inhibition and providing co-stimulation, either of which results in significantly more effective immune-mediated tumor destruction. This review will describe the pre-clinical and clinical application of antagonistic antibodies targeting the T-cell checkpoints cytotoxic T-lymphocyte antigen 4 (CTLA-4) and programmed death 1 (PD-1), and agonistic antibodies targeting the costimulatory pathways OX40 and 4-1BB. Recent progress and opportunities for future investigation of combination antibody therapy will be described.
Collapse
Affiliation(s)
- Brendan D Curti
- Earle A. Chiles Research Institute, Providence Cancer Center, 4805 NE Glisan St. 2N35, Portland, OR 97213, United States.
| | - Walter J Urba
- Earle A. Chiles Research Institute, Providence Cancer Center, 4805 NE Glisan St. 2N35, Portland, OR 97213, United States.
| |
Collapse
|
4287
|
Swaika A, Hammond WA, Joseph RW. Current state of anti-PD-L1 and anti-PD-1 agents in cancer therapy. Mol Immunol 2015; 67:4-17. [PMID: 25749122 DOI: 10.1016/j.molimm.2015.02.009] [Citation(s) in RCA: 159] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 02/05/2015] [Accepted: 02/08/2015] [Indexed: 02/07/2023]
Abstract
Immunotherapy for the treatment of cancer is rapidly evolving from therapies that globally and non-specifically simulate the immune system to more targeted activation of individual components of the immune system. The net result of this targeted approach is decreased toxicity and increased efficacy of immunotherapy. More specifically, therapies that inhibit the interaction between programmed death ligand 1 (PD-L1), present on the surface of tumor or antigen-presenting cells, and programmed death 1 (PD-1), present on the surface of activated lymphocytes, are generating much excitement and enthusiasm, even in malignancies that are not traditionally considered to be immunogenic. Herein, we review the current landscape of anti-PD-1 and anti-PD-L1 therapies in the world of oncology. We have performed a comprehensive literature search on the data available through PubMed, Medline, Scopus, the ClinicalTrials.gov registry, and abstracts from major oncology meetings in order to summarize the clinical data of anti-PD-1/PD-L1 therapies.
Collapse
Affiliation(s)
- Abhisek Swaika
- Division of Hematology and Oncology, Mayo Clinic, 4500 San Pablo Rd., Jacksonville, FL, USA
| | - William A Hammond
- Division of Hematology and Oncology, Mayo Clinic, 4500 San Pablo Rd., Jacksonville, FL, USA
| | - Richard W Joseph
- Division of Hematology and Oncology, Mayo Clinic, 4500 San Pablo Rd., Jacksonville, FL, USA.
| |
Collapse
|
4288
|
|
4289
|
Pol J, Vacchelli E, Aranda F, Castoldi F, Eggermont A, Cremer I, Sautès-Fridman C, Fucikova J, Galon J, Spisek R, Tartour E, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch: Immunogenic cell death inducers for anticancer chemotherapy. Oncoimmunology 2015; 4:e1008866. [PMID: 26137404 DOI: 10.1080/2162402x.2015.1008866] [Citation(s) in RCA: 228] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 01/14/2015] [Indexed: 02/06/2023] Open
Abstract
The term "immunogenic cell death" (ICD) is now employed to indicate a functionally peculiar form of apoptosis that is sufficient for immunocompetent hosts to mount an adaptive immune response against dead cell-associated antigens. Several drugs have been ascribed with the ability to provoke ICD when employed as standalone therapeutic interventions. These include various chemotherapeutics routinely employed in the clinic (e.g., doxorubicin, epirubicin, idarubicin, mitoxantrone, bleomycin, bortezomib, cyclophosphamide and oxaliplatin) as well as some anticancer agents that are still under preclinical or clinical development (e.g., some microtubular inhibitors of the epothilone family). In addition, a few drugs are able to convert otherwise non-immunogenic instances of cell death into bona fide ICD, and may therefore be employed as chemotherapeutic adjuvants within combinatorial regimens. This is the case of cardiac glycosides, like digoxin and digitoxin, and zoledronic acid. Here, we discuss recent developments on anticancer chemotherapy based on ICD inducers.
Collapse
Key Words
- ALL, acute lymphoblastic leukemia
- AML, acute myeloid leukemia
- CML, chronic myeloid leukemia
- DAMP, damage-associated molecular pattern
- EGFR, epidermal growth factor receptor
- EOX, epirubicin plus oxaliplatin plus capecitabine
- ER, endoplasmic reticulum
- FDA, Food and Drug Administration
- FOLFIRINOX, folinic acid plus 5-fluorouracil plus irinotecan plus oxaliplatin
- FOLFOX, folinic acid plus 5-fluorouracil plus oxaliplatin
- GEMOX, gemcitabine plus oxaliplatin
- GM-CSF, granulocyte-macrophage colony-stimulating factor
- HCC, hepatocellular carcinoma
- ICD, immunogenic cell death
- MM, multiple myeloma
- NHL, non-Hodgkin's lymphoma
- NSCLC, non-small cell lung carcinoma
- TACE, transcatheter arterial chemoembolization
- XELOX, capecitabine plus oxaliplatin
- antigen-presenting cell
- autophagy
- damage-associated molecular pattern
- dendritic cell
- endoplasmic reticulum stress
- mAb, monoclonal antibody
- type I interferon
Collapse
Affiliation(s)
- Jonathan Pol
- Gustave Roussy Cancer Campus ; Villejuif, France ; INSERM, U1138 ; Paris, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Center de Recherche des Cordeliers ; Paris, France
| | - Erika Vacchelli
- Gustave Roussy Cancer Campus ; Villejuif, France ; INSERM, U1138 ; Paris, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Center de Recherche des Cordeliers ; Paris, France
| | - Fernando Aranda
- Group of Immune receptors of the Innate and Adaptive System, Institut d'Investigacions Biomédiques August Pi i Sunyer (IDIBAPS)
| | - Francesca Castoldi
- Gustave Roussy Cancer Campus ; Villejuif, France ; INSERM, U1138 ; Paris, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Center de Recherche des Cordeliers ; Paris, France ; Faculté de Medicine; Université Paris Sud/Paris XI ; Le Kremlin-Bicêtre, France ; Sotio a.c. ; Prague, Czech Republic
| | | | - Isabelle Cremer
- INSERM, U1138 ; Paris, France ; Equipe 13, Center de Recherche des Cordeliers ; Paris, France ; Université Pierre et Marie Curie/Paris VI ; Paris, France
| | - Catherine Sautès-Fridman
- INSERM, U1138 ; Paris, France ; Equipe 13, Center de Recherche des Cordeliers ; Paris, France ; Université Pierre et Marie Curie/Paris VI ; Paris, France
| | - Jitka Fucikova
- Sotio a.c. ; Prague, Czech Republic ; Department of Immunology, 2nd Faculty of Medicine and University Hospital Motol, Charles University ; Prague, Czech Republic
| | - Jérôme Galon
- INSERM, U1138 ; Paris, France ; Université Pierre et Marie Curie/Paris VI ; Paris, France ; Laboratory of Integrative Cancer Immunology, Center de Recherche des Cordeliers ; Paris, France ; Université Paris Descartes/Paris V; Sorbonne Paris Cité ; Paris, France
| | - Radek Spisek
- Sotio a.c. ; Prague, Czech Republic ; Department of Immunology, 2nd Faculty of Medicine and University Hospital Motol, Charles University ; Prague, Czech Republic
| | - Eric Tartour
- Université Paris Descartes/Paris V; Sorbonne Paris Cité ; Paris, France ; INSERM , U970 ; Paris, France ; Paris-Cardiovascular Research Center (PARCC) ; Paris, France ; Service d'Immunologie Biologique, Hôpital Européen Georges Pompidou (HEGP); AP-HP ; Paris, France
| | - Laurence Zitvogel
- Gustave Roussy Cancer Campus ; Villejuif, France ; INSERM, U1015; CICBT507 ; Villejuif, France
| | - Guido Kroemer
- INSERM, U1138 ; Paris, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Center de Recherche des Cordeliers ; Paris, France ; Université Paris Descartes/Paris V; Sorbonne Paris Cité ; Paris, France ; Pôle de Biologie, Hôpital Européen Georges Pompidou; AP-HP ; Paris, France ; Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus ; Villejuif, France
| | - Lorenzo Galluzzi
- Gustave Roussy Cancer Campus ; Villejuif, France ; INSERM, U1138 ; Paris, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Center de Recherche des Cordeliers ; Paris, France ; Université Paris Descartes/Paris V; Sorbonne Paris Cité ; Paris, France
| |
Collapse
|
4290
|
Kroemer G, Galluzzi L. Immunotherapy of hematological cancers: PD-1 blockade for the treatment of Hodgkin's lymphoma. Oncoimmunology 2015; 4:e1008853. [PMID: 26155425 DOI: 10.1080/2162402x.2015.1008853] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Accepted: 01/14/2015] [Indexed: 10/23/2022] Open
Abstract
The blockade of immunological checkpoints has been successfully employed for the treatment of various solid neoplasms including melanoma, mesothelioma, non-small cell lung carcinoma, and renal cell carcinoma. A recent study indicates that the vast majority of patients with advanced, heavily pretreated Hodgkin's lymphoma (HL) also respond to a monoclonal antibody targeting programmed cell death 1 (PDCD1, best known as PD-1). Thus, checkpoint blockers may soon become part of our therapeutic armamentarium against hematological tumors. This would be particularly important as it would spare (at least some) patients the deleterious toxic effects of combinatorial chemotherapies and bone marrow transplantation. We anticipate that the realm of immunotherapy will eventually conquer vast portions of the territory that now belongs to hematological malignancies.
Collapse
Affiliation(s)
- Guido Kroemer
- Equipe 11 labellisée Ligue contre le Cancer; Centre de Recherche des Cordeliers ; Paris, France ; INSERM , U1138 ; Paris, France ; Université Paris Descartes/Paris V; Sorbonne Paris Cité ; Paris, France ; Université Pierre et Marie Curie/Paris VI ; Paris, France ; Metabolomics and Cell Biology Platforms; Gustave Roussy Cancer Campus ; Villejuif, France ; Pôle de Biologie; Hôpital Européen Georges Pompidou; AP-HP ; Paris, France
| | - Lorenzo Galluzzi
- Equipe 11 labellisée Ligue contre le Cancer; Centre de Recherche des Cordeliers ; Paris, France ; INSERM , U1138 ; Paris, France ; Université Paris Descartes/Paris V; Sorbonne Paris Cité ; Paris, France ; Université Pierre et Marie Curie/Paris VI ; Paris, France ; Gustave Roussy Cancer Campus ; Villejuif, France
| |
Collapse
|
4291
|
Luke JJ, Ott PA. PD-1 pathway inhibitors: the next generation of immunotherapy for advanced melanoma. Oncotarget 2015; 6:3479-92. [PMID: 25682878 PMCID: PMC4414130 DOI: 10.18632/oncotarget.2980] [Citation(s) in RCA: 126] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 12/21/2014] [Indexed: 12/20/2022] Open
Abstract
Checkpoint inhibitors are revolutionizing treatment options and expectations for patients with melanoma. Ipilimumab, a monoclonal antibody against cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4), was the first approved checkpoint inhibitor. Emerging long-term data indicate that approximately 20% of ipilimumab-treated patients achieve long-term survival. The first programmed death 1 (PD-1) inhibitor, pembrolizumab, was recently approved by the United States Food and Drug Administration for the treatment of melanoma; nivolumab was previously approved in Japan. PD-1 inhibitors are also poised to become standard of care treatment for other cancers, including non-small cell lung cancer, renal cell carcinoma and Hodgkin's lymphoma. Immunotherapy using checkpoint inhibition is a different treatment approach to chemotherapy and targeted agents: instead of directly acting on the tumor to induce tumor cell death, checkpoint inhibitors enhance or de novo stimulate antitumor immune responses to eliminate cancer cells. Initial data suggest that objective anti-tumor response rates may be higher with anti-PD-1 agents compared with ipilimumab and the safety profile may be more tolerable. This review explores the development and next steps for PD-1 pathway inhibitors, including discussion of their novel mechanism of action and clinical data to-date, with a focus on melanoma.
Collapse
Affiliation(s)
- Jason J. Luke
- Section of Hematology/Oncology, University of Chicago, Chicago, IL, USA
| | - Patrick A. Ott
- Melanoma Disease Center, Dana Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
4292
|
Ascierto PA. Combination approaches and anti-PD1 therapies: the focus of new research at ESMO and SMR. Melanoma Manag 2015; 2:9-14. [PMID: 30190825 DOI: 10.2217/mmt.14.35] [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] [Indexed: 11/21/2022] Open
Abstract
Research into the treatment of melanoma is continuing to progress, and both the European Society of Medical Oncology (26-30 September, Madrid, Spain) and the Society of Melanoma Research (13-16 November, Zurich, Switzerland) 2014 annual meetings provided important updates in this field. Areas of particular focus included combinations of a BRAF inhibitor with a MEK inhibitor (COMBI-v and CoBRIM trials) and new data on anti-PD1 therapies (nivolumab and pembrolizumab). Overall, the data reported at these two meetings again suggest that melanoma is perhaps the most dynamic field of oncology, with continuing advances in management of the disease that may improve outcomes for patients in clinical practice.
Collapse
|
4293
|
Abstract
Dysfunctional T cells can render the immune system unable to eliminate infections and cancer. Therapeutic targeting of the surface receptors that inhibit T cell function has begun to show remarkable success in clinical trials. In this Review, we discuss the potential mechanisms of action of the clinical agents that target two of these receptors, programmed cell death protein 1 (PD1) and lymphocyte activation gene 3 protein (LAG3). We also suggest correlative studies that may define the predominant mechanisms of action and identify predictive biomarkers.
Collapse
|
4294
|
|
4295
|
Maass N, Schütz F, Fasching PA, Fehm T, Janni W, Kümmel S, Kolberg HC, Lüftner D, Wallwiener M, Lux MP. Breast Cancer Update 2014 - Focus on the Patient and the Tumour. Geburtshilfe Frauenheilkd 2015; 75:170-182. [PMID: 25797960 PMCID: PMC4361168 DOI: 10.1055/s-0035-1545704] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2015] [Revised: 01/27/2015] [Accepted: 01/27/2015] [Indexed: 02/06/2023] Open
Abstract
The therapy for patients with breast cancer has developed markedly in the past ten years. Our understanding of the molecular biology of tumours and the characteristics of the patients has shaped the recent advances. In this review we present the latest knowledge about the therapy for breast cancer. There are new tests and options not only in the field of anti-HER2 therapy but also in the management of triple negative and hormone receptor-positive patients. Comprehension of prognosis and therapeutic response to chemotherapies is little by little helping to define patient groups who will not respond to chemotherapy or who do not need treatment because their prognosis is extremely good. In the field of anti-HER2 therapy, work is continuing on the development of drugs suitable for and able to overcome trastuzumab resistance. For hormone receptor-positive cancers, we now have a better understanding of which therapy groups will benefit from which anti-endocrine drugs, and which will be able to overcome a possible resistance (treatment of the PI3K pathways, inhibition of the cell cycle). Molecular tests are still being evaluated with regard to the clinical situations in which they may have the greatest relevance for therapeutic decision-making; however, evidence is also increasing as to the fields in which good predictions for the prognosis can be obtained. On the whole, more work is needed to promote our understanding of the new developments in diagnostics and therapy and to involve both physicians and patients equally in the procedures.
Collapse
Affiliation(s)
- N. Maass
- Department of Gynecology and Obstetrics, University Hospital Aachen, Aachen
| | - F. Schütz
- Frauenklinik, Universitätsklinikum Heidelberg, Heidelberg
| | - P. A. Fasching
- Department of Gynecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Comprehensive Cancer Center Erlangen-EMN, Erlangen
| | - T. Fehm
- Department of Gynecology and Obstetrics, University Hospital Düsseldorf, Heinrich Heine Universität Düsseldorf, Düsseldorf
| | - W. Janni
- Frauenklinik, Klinikum der Universität Ulm, Ulm
| | - S. Kümmel
- Brustzentrum, Kliniken Essen Mitte, Evang. Huyssens-Stiftung/Knappschaft GmbH, Essen
| | - H.-C. Kolberg
- Klinik für Gynäkologie und Geburtshilfe, Marienhospital Bottrop, Bottrop
| | - D. Lüftner
- Medizinische Klinik und Poliklinik II, Campus Charité Mitte, Berlin
| | - M. Wallwiener
- Frauenklinik, Universitätsklinikum Heidelberg, Heidelberg
| | - M. P. Lux
- Department of Gynecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Comprehensive Cancer Center Erlangen-EMN, Erlangen
| |
Collapse
|
4296
|
Zippelius A, Schreiner J, Herzig P, Müller P. Induced PD-L1 expression mediates acquired resistance to agonistic anti-CD40 treatment. Cancer Immunol Res 2015; 3:236-44. [PMID: 25623164 DOI: 10.1158/2326-6066.cir-14-0226] [Citation(s) in RCA: 105] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
CD40 stimulation on antigen-presenting cells (APC) allows direct activation of CD8(+) cytotoxic T cells, independent of CD4⁺ T-cell help. Agonistic anti-CD40 antibodies have been demonstrated to induce beneficial antitumor T-cell responses in mouse models of cancer and early clinical trials. We report here that anti-CD40 treatment induces programmed death ligand-1 (PD-L1) upregulation on tumor-infiltrating monocytes and macrophages, which was strictly dependent on T cells and IFNγ. PD-L1 expression could be counteracted by coadministration of antibodies blocking the PD-1 (programmed death-1)/PD-L1 axis as shown for T cells from tumor models and human donors. The combined treatment was highly synergistic and induced complete tumor rejection in about 50% of mice bearing MC-38 colon and EMT-6 breast tumors. Mechanistically, this was reflected by a strong increase of IFNγ and granzyme-B production in intratumoral CD8⁺ T cells. Concomitant CTLA-4 blockade further improved rejection of established tumors in mice. This study uncovers a novel mechanism of acquired resistance upon agonistic CD40 stimulation and proposes that the concomitant blockade of the PD-1/PD-L1 axis is a viable therapeutic strategy to optimize clinical outcomes.
Collapse
Affiliation(s)
- Alfred Zippelius
- Cancer Immunology and Biology, Department of Biomedicine, University Hospital Basel and University of Basel, Switzerland. Department of Medical Oncology, University Hospital Basel, Switzerland.
| | - Jens Schreiner
- Cancer Immunology and Biology, Department of Biomedicine, University Hospital Basel and University of Basel, Switzerland
| | - Petra Herzig
- Cancer Immunology and Biology, Department of Biomedicine, University Hospital Basel and University of Basel, Switzerland
| | - Philipp Müller
- Cancer Immunology and Biology, Department of Biomedicine, University Hospital Basel and University of Basel, Switzerland.
| |
Collapse
|
4297
|
Affiliation(s)
- Mario Sznol
- From the Section of Medical Oncology, Yale University School of Medicine, New Haven, CT (M.S.)
| | | |
Collapse
|
4298
|
Ascierto PA, Marincola FM. 2015: The Year of Anti-PD-1/PD-L1s Against Melanoma and Beyond. EBioMedicine 2015; 2:92-3. [PMID: 26137543 PMCID: PMC4485477 DOI: 10.1016/j.ebiom.2015.01.011] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Accepted: 01/16/2015] [Indexed: 12/03/2022] Open
Affiliation(s)
- Paolo A Ascierto
- Istituto Nazionale Tumori, Fondazione "G. Pascale", Napoli, Italy
| | | |
Collapse
|
4299
|
Spagnolo F, Ghiorzo P, Orgiano L, Pastorino L, Picasso V, Tornari E, Ottaviano V, Queirolo P. BRAF-mutant melanoma: treatment approaches, resistance mechanisms, and diagnostic strategies. Onco Targets Ther 2015; 8:157-168. [PMID: 25653539 PMCID: PMC4303458 DOI: 10.2147/ott.s39096] [Citation(s) in RCA: 110] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
BRAF inhibitors vemurafenib and dabrafenib achieved improved overall survival over chemotherapy and have been approved for the treatment of BRAF-mutated metastatic melanoma. More recently, the combination of BRAF inhibitor dabrafenib with MEK inhibitor trametinib has shown improved progression-free survival, compared to dabrafenib monotherapy, in a Phase II study and has received approval by the US Food and Drug Administration. However, even when treated with the combination, most patients develop mechanisms of acquired resistance, and some of them do not achieve tumor regression at all, because of intrinsic resistance to therapy. Along with the development of BRAF inhibitors, immunotherapy made an important step forward: ipilimumab, an anti-CTLA-4 monoclonal antibody, was approved for the treatment of metastatic melanoma; anti-PD-1 agents achieved promising results in Phase I/II trials, and data from Phase III studies will be ready soon. The availability of such drugs, which are effective regardless of BRAF status, has made the therapeutic approach more complex, as first-line treatment with BRAF inhibitors may not be the best choice for all BRAF-mutated patients. The aim of this paper is to review the systemic therapeutic options available today for patients affected by BRAF V600-mutated metastatic melanoma, as well as to summarize the mechanisms of resistance to BRAF inhibitors and discuss the possible strategies to overcome them. Moreover, since the molecular analysis of tumor specimens is now a pivotal and decisional factor in the treatment strategy of metastatic melanoma patients, the advances in the molecular detection techniques for the BRAF V600 mutation will be reported.
Collapse
Affiliation(s)
- Francesco Spagnolo
- Department of Plastic and Reconstructive Surgery, IRCCS Azienda Ospedaliera Universitaria San Martino, IST Istituto Nazionale per la Ricerca sul Cancro, Genova, Italy
| | - Paola Ghiorzo
- Department of Internal Medicine and Medical Specialties (DiMI), University of Genoa, Genova, Italy
- Genetics of Rare Cancers, IRCCS Azienda Ospedaliera Universitaria San Martino, IST Istituto Nazionale per la Ricerca sul Cancro, Genova, Italy
| | - Laura Orgiano
- Department of Medical Oncology, IRCCS Azienda Ospedaliera Universitaria San Martino, IST Istituto Nazionale per la Ricerca sul Cancro, Genova, Italy
| | - Lorenza Pastorino
- Department of Internal Medicine and Medical Specialties (DiMI), University of Genoa, Genova, Italy
- Genetics of Rare Cancers, IRCCS Azienda Ospedaliera Universitaria San Martino, IST Istituto Nazionale per la Ricerca sul Cancro, Genova, Italy
| | - Virginia Picasso
- Department of Medical Oncology, IRCCS Azienda Ospedaliera Universitaria San Martino, IST Istituto Nazionale per la Ricerca sul Cancro, Genova, Italy
| | - Elena Tornari
- Department of Medical Oncology, IRCCS Azienda Ospedaliera Universitaria San Martino, IST Istituto Nazionale per la Ricerca sul Cancro, Genova, Italy
| | - Vincenzo Ottaviano
- Department of Medical Oncology, IRCCS Azienda Ospedaliera Universitaria San Martino, IST Istituto Nazionale per la Ricerca sul Cancro, Genova, Italy
| | - Paola Queirolo
- Department of Medical Oncology, IRCCS Azienda Ospedaliera Universitaria San Martino, IST Istituto Nazionale per la Ricerca sul Cancro, Genova, Italy
| |
Collapse
|
4300
|
Callahan MK, Postow MA, Wolchok JD. CTLA-4 and PD-1 Pathway Blockade: Combinations in the Clinic. Front Oncol 2015; 4:385. [PMID: 25642417 PMCID: PMC4295550 DOI: 10.3389/fonc.2014.00385] [Citation(s) in RCA: 128] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Accepted: 12/22/2014] [Indexed: 01/29/2023] Open
Abstract
Checkpoint blocking antibodies targeting regulatory molecules on T cells such as CTLA-4 and PD-1 have reinvigorated the field of cancer immunotherapy. These agents have demonstrated clinical activity across a variety of tumor types. Now that safety and clinical activity has been demonstrated in the monotherapy setting, the field is moving in the direction of testing novel combinations.
Collapse
Affiliation(s)
- Margaret K. Callahan
- Melanoma and Immunotherapeutics Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| | - Michael A. Postow
- Melanoma and Immunotherapeutics Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| | - Jedd D. Wolchok
- Melanoma and Immunotherapeutics Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
- Ludwig Center for Cancer Immunotherapy, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| |
Collapse
|