2101
|
Chan KKW, Glenny A, Weldon JC, Furness S, Worthington HV, Wakeford H. Interventions for the treatment of oral and oropharyngeal cancers: targeted therapy and immunotherapy. Cochrane Database Syst Rev 2015; 2015:CD010341. [PMID: 26625332 PMCID: PMC9465394 DOI: 10.1002/14651858.cd010341.pub2] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Oral cancers are the sixth most common cancer worldwide, yet the prognosis following a diagnosis of oral cavity or oropharyngeal cancers remains poor, with approximately 50% survival at five years. Despite a sharp increase in research into molecularly targeted therapies and a rapid expansion in the number of clinical trials assessing new targeted therapies, their value for treating oral cancers is unclear. Therefore, it is important to summarise the evidence to determine the efficacy and toxicity of targeted therapies and immunotherapies for the treatment of these cancers. OBJECTIVES To assess the effects of molecularly targeted therapies and immunotherapies, in addition to standard therapies, for the treatment of oral cavity or oropharyngeal cancers. SEARCH METHODS We searched the following electronic databases: Cochrane Oral Health Group Trials Register (to 3 February 2015), Cochrane Central Register of Controlled Trials (CENTRAL) (The Cochrane Library, 2015, Issue 1), MEDLINE via Ovid (1946 to 3 February 2015) and EMBASE via Ovid (1980 to 3 February 2015). We searched the US National Institutes of Health Trials Register (clinicaltrials.gov), the World Health Organization Clinical Trials Registry Platform, the American Society of Clinical Oncology conference abstracts and the Radiation Therapy Oncology Group clinical trials protocols for ongoing trials. We placed no restrictions on the language or date of publication. SELECTION CRITERIA We included randomised controlled trials where more than 50% of participants had primary tumours of the oral cavity or oropharynx, and which compared targeted therapy or immunotherapy, plus standard therapy, with standard therapy alone. DATA COLLECTION AND ANALYSIS Two review authors independently screened the results of the electronic searches, extracted data and assessed the risk of bias of the included studies. We attempted to contact study authors for missing data or clarification where necessary. We combined sufficiently similar studies in meta-analyses using random-effects models when there were at least four studies and fixed-effect models when fewer than four studies. We obtained or calculated a hazard ratio (HR) and 95% confidence interval (CI) for the primary outcomes where possible. For dichotomous outcomes, we reported risk ratios (RR) and 95% CIs. MAIN RESULTS Twelve trials (2488 participants) satisfied the inclusion criteria. In the included trials, 12% of participants (298 participants) had tumours of the oral cavity and 59% (1468 participants) had oropharyngeal tumours. The remaining 29% had tumours of the larynx or hypopharynx and less than 1% had tumours at other sites.No included trial was at low risk of bias; seven had an unclear risk of bias, and five had a high risk of bias. We grouped trials by intervention type into three main comparisons: standard therapy plus epidermal growth factor receptor monoclonal antibody (EGFR mAb) therapy (follow-up period 24 to 70 months); standard therapy plus tyrosine kinase inhibitors (TKIs) (follow-up period 40 to 60 months) and standard therapy plus immunotherapy (follow-up period 24 to 70 months), all versus standard therapy alone.Moderate quality evidence showed that EGFR mAb therapy may result in 18% fewer deaths when added to standard therapy (HR of mortality 0.82; 95% CI 0.69 to 0.97; 1421 participants, three studies, 67% oropharyngeal tumours, 2% oral cavity tumours).There was also moderate quality evidence that EGFR mAb may result in 32% fewer locoregional failures when added to radiotherapy (RT) (HR 0.68; 95% CI 0.52 to 0.89; 424 participants, one study, 60% oropharyngeal tumours).A subgroup analysis separating studies by type of standard therapy (radiotherapy (RT) or chemoradiotherapy (CRT)) showed some evidence that adding EGFR mAb therapy to RT may result in a 30% reduction in the number of people whose disease progresses (HR 0.70; 95% CI 0.54 to 0.91; 424 participants, one study, 60% oropharyngeal tumours, unclear risk of bias). For the subgroup comparing EGFR mAb plus CRT with CRT alone there was insufficient evidence to determine whether adding EGFR mAb therapy to CRT impacts on progression-free survival (HR 1.08; 95% CI 0.89 to 1.32; 891 participants, one study, 70% oropharyngeal tumours, high risk of bias). The high subgroup heterogeneity meant that we were unable to pool these subgroups.There was evidence that adding cetuximab to standard therapy may result in increased skin toxicity and rash (RR 6.56; 95% CI 5.35 to 8.03; 1311 participants, two studies), but insufficient evidence to determine any difference in skin toxicity and rash in the case of nimotuzumab (RR 1.06; 95% CI 0.85 to 1.31; 92 participants, one study).There was insufficient evidence to determine whether TKIs added to standard therapy impacts on overall survival (HR 0.99; 95% CI 0.62 to 1.57; 271 participants, two studies; very low quality evidence), locoregional control (HR 0.89; 95% CI 0.53 to 1.49; 271 participants, two studies; very low quality evidence), disease-free survival (HR 1.51; 95% CI 0.61 to 3.71; 60 participants, one study; very low quality evidence) or progression-free survival (HR 0.80; 95% CI 0.51 to 1.28; 271 participants, two studies; very low quality evidence). We did find evidence of an increase in skin rash (erlotinib: RR 6.57; 95% CI 3.60 to 12.00; 191 participants, one study; lapatinib: RR 2.02; 95% CI 1.23 to 3.32; 67 participants, one study) and gastrointestinal complaints (lapatinib: RR 15.53; 95% CI 2.18 to 110.55; 67 participants, one study).We found very low quality evidence from one small trial that adding recombinant interleukin (rIL-2) to surgery may increase overall survival (HR 0.52; 95% CI 0.31 to 0.87; 201 participants, 62% oral cavity tumours, 38% oropharyngeal tumours) and there was insufficient evidence to determine whether rIL-2 impacts on adverse effects. AUTHORS' CONCLUSIONS We found some evidence that adding EGFR mAb to standard therapy may increase overall survival, progression-free survival and locoregional control, while resulting in an increase in skin toxicity for some mAb (cetuximab).There is insufficient evidence to determine whether adding TKIs to standard therapies changes any of our primary outcomes.Very low quality evidence from a single study suggests that rIL-2 combined with surgery may increase overall survival compared with surgery alone.
Collapse
Affiliation(s)
- Kelvin KW Chan
- Sunnybrook Health Sciences CentreSunnybrook Odette Cancer Centre2075 Bayview AvenueTorontoONCanadaM4N3M5
| | - Anne‐Marie Glenny
- School of Dentistry, The University of ManchesterCochrane Oral Health GroupJR Moore BuildingOxford RoadManchesterUKM13 9PL
| | - Jo C Weldon
- School of Dentistry, The University of ManchesterCochrane Oral Health GroupJR Moore BuildingOxford RoadManchesterUKM13 9PL
| | - Susan Furness
- School of Dentistry, The University of ManchesterCochrane Oral Health GroupJR Moore BuildingOxford RoadManchesterUKM13 9PL
| | - Helen V Worthington
- School of Dentistry, The University of ManchesterCochrane Oral Health GroupJR Moore BuildingOxford RoadManchesterUKM13 9PL
| | - Helen Wakeford
- School of Dentistry, The University of ManchesterCochrane Oral Health GroupJR Moore BuildingOxford RoadManchesterUKM13 9PL
| | | |
Collapse
|
2102
|
Abstract
Advanced hepatocellular carcinoma (HCC) is a serious therapeutic challenge and targeted therapies only provide a modest benefit in terms of overall survival. Novel approaches are urgently needed for the treatment of this prevalent malignancy. Evidence demonstrating the antigenicity of tumour cells, the discovery that immune checkpoint molecules have an essential role in immune evasion of tumour cells, and the impressive clinical results achieved by blocking these inhibitory receptors, are revolutionizing cancer immunotherapy. Here, we review the data on HCC immunogenicity, the mechanisms for HCC immune subversion and the different immunotherapies that have been tested to treat HCC. Taking into account the multiplicity of hyperadditive immunosuppressive forces acting within the HCC microenvironment, a combinatorial approach is advised. Strategies include combinations of systemic immunomodulation and gene therapy, cell therapy or virotherapy.
Collapse
|
2103
|
Naidoo J, Page DB, Li BT, Connell LC, Schindler K, Lacouture ME, Postow MA, Wolchok JD. Toxicities of the anti-PD-1 and anti-PD-L1 immune checkpoint antibodies. Ann Oncol 2015; 26:2375-91. [PMID: 26371282 PMCID: PMC6267867 DOI: 10.1093/annonc/mdv383] [Citation(s) in RCA: 1051] [Impact Index Per Article: 105.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 09/01/2015] [Indexed: 12/17/2022] Open
Abstract
Immune checkpoint antibodies that augment the programmed cell death protein 1 (PD-1)/PD-L1 pathway have demonstrated antitumor activity across multiple malignancies, and gained recent regulatory approval as single-agent therapy for the treatment of metastatic malignant melanoma and nonsmall-cell lung cancer. Knowledge of toxicities associated with PD-1/PD-L1 blockade, as well as effective management algorithms for these toxicities, is pivotal in order to optimize clinical efficacy and safety. In this article, we review selected published and presented clinical studies investigating single-agent anti-PD-1/PD-L1 therapy and trials of combination approaches with other standard anticancer therapies, in multiple tumor types. We summarize the key adverse events reported in these studies and their management algorithms.
Collapse
Affiliation(s)
- J Naidoo
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore
| | - D B Page
- Providence Portland Medical Center and Earl A. Chiles Research Institute, Portland
| | - B T Li
- Department of Medicine and Ludwig Center, Memorial Sloan Kettering Cancer Center, New York, USA
| | - L C Connell
- Department of Medicine and Ludwig Center, Memorial Sloan Kettering Cancer Center, New York, USA
| | - K Schindler
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - M E Lacouture
- Dermatology Service, Memorial Sloan Kettering Cancer Center, New York Department of Medicine, Weill Cornell Medical College, New York, USA
| | - M A Postow
- Department of Medicine and Ludwig Center, Memorial Sloan Kettering Cancer Center, New York, USA Department of Medicine, Weill Cornell Medical College, New York, USA
| | - J D Wolchok
- Department of Medicine and Ludwig Center, Memorial Sloan Kettering Cancer Center, New York, USA Department of Medicine, Weill Cornell Medical College, New York, USA
| |
Collapse
|
2104
|
van Rooijen JM, Stutvoet TS, Schröder CP, de Vries EG. Immunotherapeutic options on the horizon in breast cancer treatment. Pharmacol Ther 2015; 156:90-101. [DOI: 10.1016/j.pharmthera.2015.09.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
2105
|
|
2106
|
Hammerich L, Binder A, Brody JD. In situ vaccination: Cancer immunotherapy both personalized and off-the-shelf. Mol Oncol 2015; 9:1966-81. [PMID: 26632446 PMCID: PMC5528727 DOI: 10.1016/j.molonc.2015.10.016] [Citation(s) in RCA: 136] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Revised: 10/20/2015] [Accepted: 10/20/2015] [Indexed: 01/15/2023] Open
Abstract
As cancer immunotherapy continues to benefit from novel approaches which cut immune 'brake pedals' (e.g. anti-PD1 and anti-CTLA4 antibodies) and push immune cell gas pedals (e.g. IL2, and IFNα) there will be increasing need to develop immune 'steering wheels' such as vaccines to guide the immune system specifically toward tumor associated antigens. Two primary hurdles in cancer vaccines have been: identification of universal antigens to be used in 'off-the-shelf' vaccines for common cancers, and 2) logistical hurdles of ex vivo production of individualized whole tumor cell vaccines. Here we summarize approaches using 'in situ vaccination' in which intratumoral administration of off-the-shelf immunomodulators have been developed to specifically induce (or amplify) T cell responses to each patient's individual tumor. Clinical studies have confirmed the induction of systemic immune and clinical responses to such approaches and preclinical models have suggested ways to further potentiate the translation of in situ vaccine trials for our patients.
Collapse
Affiliation(s)
- Linda Hammerich
- Icahn School of Medicine at Mount Sinai Hess Center for Science and Medicine, United States
| | - Adam Binder
- Icahn School of Medicine at Mount Sinai Hess Center for Science and Medicine, United States
| | - Joshua D Brody
- Icahn School of Medicine at Mount Sinai Hess Center for Science and Medicine, United States.
| |
Collapse
|
2107
|
Duan L, Mukherjee EM, Narayan D. Tailoring the Treatment of Melanoma: Implications for Personalized Medicine. THE YALE JOURNAL OF BIOLOGY AND MEDICINE 2015; 88:389-95. [PMID: 26604863 PMCID: PMC4654188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Oncology has been revolutionized by the ability to selectively inhibit the growth of cancerous cells while ostensibly avoiding the disruption of proteins and pathways necessary for normal cellular function. This paradigm has triggered an explosion of targeted therapies for cancer, creating a burgeoning billion-dollar industry of small molecules and monoclonal antibodies [1]. Largely due to these new treatments, spending on cancer pharmaceuticals has surpassed $100 billion worldwide [2]. In particular, the treatment of melanoma, a deadly and fast-spreading form of skin cancer, has been transformed by these new targeted therapies. In this mini-review, we summarize the progress made in the field of personalized treatment of melanoma, with an emphasis on targeted therapies. We then outline future directions for treatment, including novel cell-mediated therapies and new potential targets.
Collapse
Affiliation(s)
- Linna Duan
- Yale School of Medicine, New Haven, Connecticut
| | - Eric M. Mukherjee
- Department of Dermatology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Deepak Narayan
- Department of Plastic and Reconstructive Surgery, Yale School of Medicine, New Haven, Connecticut
| |
Collapse
|
2108
|
Márquez-Rodas I, Cerezuela P, Soria A, Berrocal A, Riso A, González-Cao M, Martín-Algarra S. Immune checkpoint inhibitors: therapeutic advances in melanoma. ANNALS OF TRANSLATIONAL MEDICINE 2015; 3:267. [PMID: 26605313 DOI: 10.3978/j.issn.2305-5839.2015.10.27] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In recent years, new strategies for treating melanoma have been introduced, improving the outlook for this challenging disease. One of the most important advances has been the development of immunotherapy. The better understanding of the role of the immunological system in tumor control has paved the way for strategies to enhance the immune response against cancer cells. Monoclonal antibodies (mAbs) against the immune checkpoints cytotoxic T-lymphocyte antigen-4 (CTLA-4) and programmed cell death protein 1 (PD-1) and its ligand (PD-L1) have demonstrated high activity in melanoma and other tumors. Ipilimumab, an anti CTLA-4 antibody, was the first drug of this class that was approved. Although the response rate with ipilimumab is low (less than 20% of patients have objective responses), 20% of patients have long survival, with similar results in the first and second line settings. Nivolumab and pembrolizumab, both anti PD-1 inhibitors, have been approved for the treatment of melanoma, with response rates of 40% and a demonstrated survival advantage in phase III trials. This has marked a new era in the treatment of metastatic melanoma and much research is now ongoing with other drugs targeting checkpoint inhibitors. In addition, the agonist of activating molecules on T cells and their combinations are being investigated. Herein we review the clinical development of checkpoint inhibitors and their approval for treatment of metastatic melanoma.
Collapse
Affiliation(s)
- Ivan Márquez-Rodas
- 1 Servicio de Oncología Médica, Hospital General Universitario Gregorio Marañon, Madrid, Spain ; 2 Spanish Melanoma Group (GEM); 3 Servicio Oncología Médica, Hospital General Universitario Santa Lucía, Cartagena, Spain ; 4 Servicio de Oncología Médica, Universitario Ramon y Cajal, Madrid, Spain ; 5 Servicio de Oncología Médica, Hospital General Universitario de Valencia, Valencia, Spain ; 6 Translational Cancer Research Unit, Instituto Oncológico Dr Rosell, Quirón Dexeus University Hospital, Barcelona, Spain ; 7 Servicio de Oncología Médica, Clínica Universitaria de Navarra, Pamplona, Spain
| | - Pablo Cerezuela
- 1 Servicio de Oncología Médica, Hospital General Universitario Gregorio Marañon, Madrid, Spain ; 2 Spanish Melanoma Group (GEM); 3 Servicio Oncología Médica, Hospital General Universitario Santa Lucía, Cartagena, Spain ; 4 Servicio de Oncología Médica, Universitario Ramon y Cajal, Madrid, Spain ; 5 Servicio de Oncología Médica, Hospital General Universitario de Valencia, Valencia, Spain ; 6 Translational Cancer Research Unit, Instituto Oncológico Dr Rosell, Quirón Dexeus University Hospital, Barcelona, Spain ; 7 Servicio de Oncología Médica, Clínica Universitaria de Navarra, Pamplona, Spain
| | - Ainara Soria
- 1 Servicio de Oncología Médica, Hospital General Universitario Gregorio Marañon, Madrid, Spain ; 2 Spanish Melanoma Group (GEM); 3 Servicio Oncología Médica, Hospital General Universitario Santa Lucía, Cartagena, Spain ; 4 Servicio de Oncología Médica, Universitario Ramon y Cajal, Madrid, Spain ; 5 Servicio de Oncología Médica, Hospital General Universitario de Valencia, Valencia, Spain ; 6 Translational Cancer Research Unit, Instituto Oncológico Dr Rosell, Quirón Dexeus University Hospital, Barcelona, Spain ; 7 Servicio de Oncología Médica, Clínica Universitaria de Navarra, Pamplona, Spain
| | - Alfonso Berrocal
- 1 Servicio de Oncología Médica, Hospital General Universitario Gregorio Marañon, Madrid, Spain ; 2 Spanish Melanoma Group (GEM); 3 Servicio Oncología Médica, Hospital General Universitario Santa Lucía, Cartagena, Spain ; 4 Servicio de Oncología Médica, Universitario Ramon y Cajal, Madrid, Spain ; 5 Servicio de Oncología Médica, Hospital General Universitario de Valencia, Valencia, Spain ; 6 Translational Cancer Research Unit, Instituto Oncológico Dr Rosell, Quirón Dexeus University Hospital, Barcelona, Spain ; 7 Servicio de Oncología Médica, Clínica Universitaria de Navarra, Pamplona, Spain
| | - Aldo Riso
- 1 Servicio de Oncología Médica, Hospital General Universitario Gregorio Marañon, Madrid, Spain ; 2 Spanish Melanoma Group (GEM); 3 Servicio Oncología Médica, Hospital General Universitario Santa Lucía, Cartagena, Spain ; 4 Servicio de Oncología Médica, Universitario Ramon y Cajal, Madrid, Spain ; 5 Servicio de Oncología Médica, Hospital General Universitario de Valencia, Valencia, Spain ; 6 Translational Cancer Research Unit, Instituto Oncológico Dr Rosell, Quirón Dexeus University Hospital, Barcelona, Spain ; 7 Servicio de Oncología Médica, Clínica Universitaria de Navarra, Pamplona, Spain
| | - María González-Cao
- 1 Servicio de Oncología Médica, Hospital General Universitario Gregorio Marañon, Madrid, Spain ; 2 Spanish Melanoma Group (GEM); 3 Servicio Oncología Médica, Hospital General Universitario Santa Lucía, Cartagena, Spain ; 4 Servicio de Oncología Médica, Universitario Ramon y Cajal, Madrid, Spain ; 5 Servicio de Oncología Médica, Hospital General Universitario de Valencia, Valencia, Spain ; 6 Translational Cancer Research Unit, Instituto Oncológico Dr Rosell, Quirón Dexeus University Hospital, Barcelona, Spain ; 7 Servicio de Oncología Médica, Clínica Universitaria de Navarra, Pamplona, Spain
| | - Salvador Martín-Algarra
- 1 Servicio de Oncología Médica, Hospital General Universitario Gregorio Marañon, Madrid, Spain ; 2 Spanish Melanoma Group (GEM); 3 Servicio Oncología Médica, Hospital General Universitario Santa Lucía, Cartagena, Spain ; 4 Servicio de Oncología Médica, Universitario Ramon y Cajal, Madrid, Spain ; 5 Servicio de Oncología Médica, Hospital General Universitario de Valencia, Valencia, Spain ; 6 Translational Cancer Research Unit, Instituto Oncológico Dr Rosell, Quirón Dexeus University Hospital, Barcelona, Spain ; 7 Servicio de Oncología Médica, Clínica Universitaria de Navarra, Pamplona, Spain
| |
Collapse
|
2109
|
Smyth MJ, Ngiow SF, Ribas A, Teng MWL. Combination cancer immunotherapies tailored to the tumour microenvironment. Nat Rev Clin Oncol 2015; 13:143-58. [PMID: 26598942 DOI: 10.1038/nrclinonc.2015.209] [Citation(s) in RCA: 698] [Impact Index Per Article: 69.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Evidence suggests that cancer immunotherapy will be a major part of the combination treatment plan for many patients with many cancer types in the near future. There are many types of immune processes involving different antitumour and tumour-promoting leucocytes, and tumour cells use many strategies to evade the immune response. The tumour microenvironment can help determine which immune suppressive pathways become activated to restrain antitumour immunity. This includes immune checkpoint receptors on effector T-cells and myeloid cells, and release of inhibitory cytokines and metabolites. Therapeutic approaches that target these pathways, particularly immune-checkpoint receptors, can induce durable antitumour responses in patients with advanced-stage cancers, including melanoma. Nevertheless, many patients do not have a good response to monotherapy approaches and alternative strategies are required to achieve optimal therapeutic benefit. These strategies include eliminating the bulk of tumour cells to provoke tumour-antigen release and antigen-presenting cell (APC) function, using adjuvants to enhance APC function, and using agents that enhance effector-cell activity. In this Review, we discuss the stratification of the tumour microenvironment according to tumour-infiltrating lymphocytes and PD-L1 expression in the tumour, and how this stratification enables the design of optimal combination cancer therapies tailored to target different tumour microenvironments.
Collapse
Affiliation(s)
- Mark J Smyth
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston 4006, Queensland, Australia
| | - Shin Foong Ngiow
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston 4006, Queensland, Australia
| | - Antoni Ribas
- Department of Medicine, Division of Haematology/Oncology, Jonsson Comprehensive Cancer Center at UCLA, Los Angeles, California 90095, USA
| | - Michele W L Teng
- Cancer Immunoregulation and Immunotherapy Laboratory QIMR Berghofer Medical Research Institute, Herston, 4006 Queensland, Australia
| |
Collapse
|
2110
|
Abstract
Radiation therapy (RT) is a cornerstone in oncologic management and is employed in various curative and palliative scenarios for local-regional control. RT is thought to locally control tumor cells by direct physical DNA damage or indirect insults from reactive oxygen species. Therapeutic effects apart from those observed at the treatment target, that is, abscopal effect, have been observed for several decades, though the underlying mechanisms regulating this phenomenon have been unclear. Accumulating evidence now suggests that the immune system is a major determinant in regulating the abscopal effect. It is now evident that RT may also enhance immunologic responses to tumors by creating an in situ vaccine by eliciting antigen release from dying tumor cells. Harnessing the specificity and dynamic nature of the immune system to target tumors in conjunction with RT is an emerging field with much promise. To optimize this approach, it is important to systematically evaluate the intricacies of the host immune system, the new generation of immunotherapeutics and the RT approach. Here we will discuss the current biologic mechanisms thought to regulate the RT-induced abscopal effect and how these may be translated to the clinical setting.
Collapse
|
2111
|
|
2112
|
Huang Z, Li H, Fan Y. [Clinical Research Progress of Anti PD-1/PD-L1 Monoclonal Antibody in the Treatment of Lung Cancer]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2015; 18:706-13. [PMID: 26582228 PMCID: PMC6000315 DOI: 10.3779/j.issn.1009-3419.2015.11.09] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
近期,抗程序性死亡分子1(programmed death-1, PD-1) /PD-1配体(PD-1 ligand, PD-L1)单抗在晚期非小细胞肺癌的治疗中纷纷取得了突破性进展,相关研究迅速覆盖到肺癌的其他领域,如小细胞肺癌、局部晚期非小细胞肺癌;并且开始尝试与现有治疗手段的各种组合,如联合化疗、靶向药物及放疗等。但是,仍有许多问题有待解决,如寻找理想的预测疗效的生物标记物、探索不同的联合治疗模式、明确原发及继发耐药的机制等等。随着这些问题的相继解决,抗PD-1/PD-L1单抗在肺癌的治疗中将会有更加广阔的应用空间。
Collapse
Affiliation(s)
- Zhiyu Huang
- Department of Thoracic Medical Oncology, Zhejiang Cancer Hospital, Hangzhou 310022, China
| | - Hui Li
- Department of Thoracic Medical Oncology, Zhejiang Cancer Hospital, Hangzhou 310022, China
| | - Yun Fan
- Department of Thoracic Medical Oncology, Zhejiang Cancer Hospital, Hangzhou 310022, China
| |
Collapse
|
2113
|
Cang S, Iragavarapu C, Savooji J, Song Y, Liu D. ABT-199 (venetoclax) and BCL-2 inhibitors in clinical development. J Hematol Oncol 2015; 8:129. [PMID: 26589495 PMCID: PMC4654800 DOI: 10.1186/s13045-015-0224-3] [Citation(s) in RCA: 207] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 11/10/2015] [Indexed: 12/21/2022] Open
Abstract
With the advent of new agents targeting CD20, Bruton's tyrosine kinase, and phosphoinositol-3 kinase for chronic lymphoid leukemia (CLL), more treatment options exist than ever before. B-cell lymphoma-2 (BCL-2) plays a major role in cellular apoptosis and is a druggable target. Small molecule inhibitors of BCL-2 are in active clinical studies. ABT-199 (venetoclax, RG7601, GDC-0199) has been granted breakthrough designation by FDA for relapsed or refractory CLL with 17p deletion. In this review, we summarized the latest clinical development of ABT-199/venetoclax and other novel agents targeting the BCL-2 proteins.
Collapse
Affiliation(s)
- Shundong Cang
- Department of Oncology, The Henan Province People's Hospital, Zhengzhou, China
| | - Chaitanya Iragavarapu
- Department of Medicine, Westchester Medical Center and New York Medical College, Valhalla, New York, 10595, USA
| | - John Savooji
- Department of Medicine, Westchester Medical Center and New York Medical College, Valhalla, New York, 10595, USA
| | - Yongping Song
- Henan Cancer Hospital and the Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | - Delong Liu
- Henan Cancer Hospital and the Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
2114
|
Hebeisen M, Allard M, Gannon PO, Schmidt J, Speiser DE, Rufer N. Identifying Individual T Cell Receptors of Optimal Avidity for Tumor Antigens. Front Immunol 2015; 6:582. [PMID: 26635796 PMCID: PMC4649060 DOI: 10.3389/fimmu.2015.00582] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 10/30/2015] [Indexed: 02/02/2023] Open
Abstract
Cytotoxic T cells recognize, via their T cell receptors (TCRs), small antigenic peptides presented by the major histocompatibility complex (pMHC) on the surface of professional antigen-presenting cells and infected or malignant cells. The efficiency of T cell triggering critically depends on TCR binding to cognate pMHC, i.e., the TCR–pMHC structural avidity. The binding and kinetic attributes of this interaction are key parameters for protective T cell-mediated immunity, with stronger TCR–pMHC interactions conferring superior T cell activation and responsiveness than weaker ones. However, high-avidity TCRs are not always available, particularly among self/tumor antigen-specific T cells, most of which are eliminated by central and peripheral deletion mechanisms. Consequently, systematic assessment of T cell avidity can greatly help distinguishing protective from non-protective T cells. Here, we review novel strategies to assess TCR–pMHC interaction kinetics, enabling the identification of the functionally most-relevant T cells. We also discuss the significance of these technologies in determining which cells within a naturally occurring polyclonal tumor-specific T cell response would offer the best clinical benefit for use in adoptive therapies, with or without T cell engineering.
Collapse
Affiliation(s)
- Michael Hebeisen
- Department of Oncology, Lausanne University Hospital Center (CHUV), University of Lausanne , Epalinges , Switzerland
| | - Mathilde Allard
- Department of Oncology, Lausanne University Hospital Center (CHUV), University of Lausanne , Epalinges , Switzerland
| | - Philippe O Gannon
- Department of Oncology, Lausanne University Hospital Center (CHUV), University of Lausanne , Epalinges , Switzerland
| | - Julien Schmidt
- Ludwig Center for Cancer Research, University of Lausanne , Epalinges , Switzerland ; TCMetrix Sàrl , Epalinges , Switzerland
| | - Daniel E Speiser
- Department of Oncology, Lausanne University Hospital Center (CHUV), University of Lausanne , Epalinges , Switzerland ; Ludwig Center for Cancer Research, University of Lausanne , Epalinges , Switzerland
| | - Nathalie Rufer
- Department of Oncology, Lausanne University Hospital Center (CHUV), University of Lausanne , Epalinges , Switzerland ; Ludwig Center for Cancer Research, University of Lausanne , Epalinges , Switzerland
| |
Collapse
|
2115
|
Metastatic melanoma treatment: Combining old and new therapies. Crit Rev Oncol Hematol 2015; 98:242-53. [PMID: 26616525 DOI: 10.1016/j.critrevonc.2015.11.011] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Revised: 10/16/2015] [Accepted: 11/12/2015] [Indexed: 01/04/2023] Open
Abstract
Metastatic melanoma is an aggressive form of cancer characterised by poor prognosis and a complex etiology. Until 2010, the treatment options for metastatic melanoma were very limited. Largely ineffective dacarbazine, temozolamide or fotemustine were the only agents in use for 35 years. In recent years, the development of molecularly targeted inhibitors in parallel with the development of checkpoint inhibition immunotherapies has rapidly improved the outcomes for metastatic melanoma patients. Despite these new therapies showing initial promise; resistance and poor duration of response have limited their effectiveness as monotherapies. Here we provide an overview of the history of melanoma treatment, as well as the current treatments in development. We also discuss the future of melanoma treatment as we go beyond monotherapies to a combinatorial approach. Combining older therapies with the new molecular and immunotherapies will be the most promising way forward for treatment of metastatic melanoma.
Collapse
|
2116
|
Abstract
INTRODUCTION Diversion of the immune checkpoint PD-1/PD-L1 by a tumor in order to escape antitumor immunity is a hallmark of NSCLC, but offers promising new strategies. Nivolumab, a fully human monoclonal antibody, is the first PD-1 inhibitor to be approved to treat metastatic NSCLC after exciting results obtained from clinical trials. AREAS COVERED This review aims to:) clarify the mechanism of action and toxicities of PD-1 inhibitors; recapitulate the results from various clinical trials that have evaluated nivolumab as a monotherapy for metastatic NSCLC; discuss the clinical and translational research axes to better use this molecule; and summarize the therapeutic combinations currently under evaluation. EXPERT OPINION The contribution of this molecule to treat NSCLC is undeniable, making it a new standard of care after prior chemotherapy. Its toxicity profile is favorable but a good knowledge of new and potentially severe immune-related adverse effects such as endocrinopathy or interstitial pneumonitis is essential for its early detection and management. Better selection of patients is needed, particularly based on the discovery of predictive biomarkers, such as PD-L1 expression. Multiple associations with other checkpoint inhibitors, chemotherapy and targeted therapies are currently being studied and should pave the way toward new uses for this drug.
Collapse
Affiliation(s)
- Nicolas Guibert
- a Centre Hospitalier Universitaire, Thoracic Oncology Unit, Respiratory Disease Department , Hôpital Larrey, Université Paul Sabatier , CHU Toulouse, Chemin de Pouvourville, Toulouse , Toulouse Cedex 31059 , France
| | - Julien Mazières
- a Centre Hospitalier Universitaire, Thoracic Oncology Unit, Respiratory Disease Department , Hôpital Larrey, Université Paul Sabatier , CHU Toulouse, Chemin de Pouvourville, Toulouse , Toulouse Cedex 31059 , France.,b Hôpital Larrey , Institut Universitaire du Cancer , Toulouse , Toulouse Cedex 31059 , France
| |
Collapse
|
2117
|
Postel-Vinay S, Aspeslagh S, Lanoy E, Robert C, Soria JC, Marabelle A. Challenges of phase 1 clinical trials evaluating immune checkpoint-targeted antibodies. Ann Oncol 2015; 27:214-24. [PMID: 26578728 DOI: 10.1093/annonc/mdv550] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 10/27/2015] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Immunostimulatory monoclonal antibodies (imAbs) targeting immune checkpoint molecules are revolutionizing oncology not only regarding cancer therapeutics and clinical care, but also from a drug development point of view. A handful of first-generation molecules have been approved so far based on their tremendous efficacy, after an expedited development phase that has challenged most paradigms established in the era of conventional cytotoxic therapy and to some extent molecularly targeted agents. A huge wave of second-generation imAbs is just entering into phase 1 trials now, in monotherapy or in combination. In order to maximize their chances of success in early phase trials, and eventually for patients' benefit, their clinical development has to benefit from lessons learnt from previous imAbs phase 1 trials. MATERIALS AND METHODS We reviewed the early clinical development of anti-cytotoxic T-lymphocyte antigen 4 and anti-programmed death-1 receptor/ligand. Particularities of each agent, including safety, dose--toxicity and dose--efficacy relationships, scheduling, pharmacokinetics (PK), pharmacodynamics (PD), trial design, biomarkers, response assessment and overall drug development strategies, are described and challenged. RESULTS As opposed to conventional cytotoxic agents, dose of imAbs is not linearly associated with efficacy and toxicity. Therefore, the definition of a minimal immunologically active dose could be proposed. Traditional patient eligibility criteria might also be revisited as the toxicity profile and mechanism of toxicity--immune-related adverse events--are mostly known and their physiopathology somehow less unexpected than with molecularly targeted small molecules. Most challenging are the comprehensive investigation of complex PK and PD characteristics as well as the definition of patient selection biomarkers. Finally, the early focus on efficacy (and not only dose confirmation) in expansion cohorts challenges the traditional phase 1/2/3 drug development process. CONCLUSION Several drug development paradigms have been challenged by imAbs. Here, we discuss novel approaches for an efficient and successful drug development of these agents.
Collapse
Affiliation(s)
- S Postel-Vinay
- DITEP (Département d'Innovations Thérapeutiques et Essais Précoces), Gustave Roussy, Villejuif Faculty of Medicine, Université Paris Saclay, Université Paris-Sud, Paris Inserm Unit U981, Gustave Roussy, Villejuif
| | - S Aspeslagh
- DITEP (Département d'Innovations Thérapeutiques et Essais Précoces), Gustave Roussy, Villejuif
| | - E Lanoy
- Biostatistics and Epidemiology Unit, Gustave-Roussy, Villejuif Inserm Unit U1018, CESP, Université Paris-Sud, Université Paris-Saclay, Villejuif
| | - C Robert
- Faculty of Medicine, Université Paris Saclay, Université Paris-Sud, Paris Inserm Unit U981, Gustave Roussy, Villejuif Department of Medical Oncology, Gustave Roussy, Villejuif
| | - J-C Soria
- DITEP (Département d'Innovations Thérapeutiques et Essais Précoces), Gustave Roussy, Villejuif Faculty of Medicine, Université Paris Saclay, Université Paris-Sud, Paris Inserm Unit U981, Gustave Roussy, Villejuif
| | - A Marabelle
- DITEP (Département d'Innovations Thérapeutiques et Essais Précoces), Gustave Roussy, Villejuif Inserm Unit U1015, Gustave Roussy, Villejuif, France
| |
Collapse
|
2118
|
Rajani KR, Vile RG. Harnessing the Power of Onco-Immunotherapy with Checkpoint Inhibitors. Viruses 2015; 7:5889-901. [PMID: 26580645 PMCID: PMC4664987 DOI: 10.3390/v7112914] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 10/26/2015] [Accepted: 10/29/2015] [Indexed: 12/11/2022] Open
Abstract
Oncolytic viruses represent a diverse class of replication competent viruses that curtail tumor growth. These viruses, through their natural ability or through genetic modifications, can selectively replicate within tumor cells and induce cell death while leaving normal cells intact. Apart from the direct oncolytic activity, these viruses mediate tumor cell death via the induction of innate and adaptive immune responses. The field of oncolytic viruses has seen substantial advancement with the progression of numerous oncolytic viruses in various phases of clinical trials. Tumors employ a plethora of mechanisms to establish growth and subsequently metastasize. These include evasion of immune surveillance by inducing up-regulation of checkpoint proteins which function to abrogate T cell effector functions. Currently, antibodies blocking checkpoint proteins such as anti-cytotoxic T-lymphocyte antigen-4 (CTLA-4) and anti-programmed cell death-1 (PD-1) have been approved to treat cancer and shown to impart durable clinical responses. These antibodies typically need pre-existing active immune tumor microenvironment to establish durable clinical outcomes and not every patient responds to these therapies. This review provides an overview of published pre-clinical studies demonstrating superior therapeutic efficacy of combining oncolytic viruses with checkpoint blockade compared to monotherapies. These studies provide compelling evidence that oncolytic therapy can be potentiated by coupling it with checkpoint therapies.
Collapse
Affiliation(s)
- Karishma R Rajani
- Department of Molecular Medicine; Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA.
| | - Richard G Vile
- Department of Molecular Medicine; Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA.
- Department of Immunology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA.
| |
Collapse
|
2119
|
Spain L, Younger E, Hatipoglu E, Larkin J. Pembrolizumab in the management of metastatic melanoma. Melanoma Manag 2015; 2:315-325. [PMID: 30190860 DOI: 10.2217/mmt.15.33] [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: 12/12/2022] Open
Abstract
Pembrolizumab is a humanized IgG4 anti-PD-1 antibody that plays a major role in the treatment of advanced melanoma. Through blockade of PD-1, it leads to an increase in effector T-cell activity in the tumor microenvironment. Clinical trial outcomes for pembrolizumab in addition to pharmacokinetics, pharmacodynamics and safety of the compound are discussed in this article. Phase I trials have demonstrated safety and efficacy of pembrolizumab in advanced, pretreated melanoma patients. When compared with chemotherapy in a Phase II trial of ipilimumab-refractory patients, those treated with pembrolizumab showed superior progression-free survival. In addition, in the pivotal Phase III trial pembrolizumab improved overall survival compared with ipilimumab in patients naive to immune checkpoint inhibition. Pembrolizumab is well tolerated and has a favorable safety profile. Common adverse events are fatigue, rash, itching and diarrhea. Less frequent immune-related adverse events include hypothyroidism, colitis, hepatitis and pneumonitis.
Collapse
Affiliation(s)
- Lavinia Spain
- The Royal Marsden Hospital, Fulham Road, London, SW3 6JJ, UK
| | - Eugenie Younger
- The Royal Marsden Hospital, Fulham Road, London, SW3 6JJ, UK
| | - Emine Hatipoglu
- The Royal Marsden Hospital, Fulham Road, London, SW3 6JJ, UK
| | - James Larkin
- The Royal Marsden Hospital, Fulham Road, London, SW3 6JJ, UK
| |
Collapse
|
2120
|
Chae YK, Chung SY, Davis AA, Carneiro BA, Chandra S, Kaplan J, Kalyan A, Giles FJ. Adenoid cystic carcinoma: current therapy and potential therapeutic advances based on genomic profiling. Oncotarget 2015; 6:37117-34. [PMID: 26359351 PMCID: PMC4741919 DOI: 10.18632/oncotarget.5076] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 08/08/2015] [Indexed: 11/25/2022] Open
Abstract
Adenoid cystic carcinoma (ACC) is a rare cancer with high potential for recurrence and metastasis. Efficacy of current treatment options, particularly for advanced disease, is very limited. Recent whole genome and exome sequencing has dramatically improved our understanding of ACC pathogenesis. A balanced translocation resulting in the MYB-NFIB fusion gene appears to be a fundamental signature of ACC. In addition, sequencing has identified a number of other driver genes mutated in downstream pathways common to other well-studied cancers. Overexpression of oncogenic proteins involved in cell growth, adhesion, cell cycle regulation, and angiogenesis are also present in ACC. Collectively, studies have identified genes and proteins for targeted, mechanism-based, therapies based on tumor phenotypes, as opposed to nonspecific cytotoxic agents. In addition, although few studies in ACC currently exist, immunotherapy may also hold promise. Better genetic understanding will enable treatment with novel targeted agents and initial exploration of immune-based therapies with the goal of improving outcomes for patients with ACC.
Collapse
Affiliation(s)
- Young Kwang Chae
- Northwestern Medicine Developmental Therapeutics Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Su Yun Chung
- Northwestern Medicine Developmental Therapeutics Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Andrew A. Davis
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Benedito A. Carneiro
- Northwestern Medicine Developmental Therapeutics Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Sunandana Chandra
- Northwestern Medicine Developmental Therapeutics Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Jason Kaplan
- Northwestern Medicine Developmental Therapeutics Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Aparna Kalyan
- Northwestern Medicine Developmental Therapeutics Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Francis J. Giles
- Northwestern Medicine Developmental Therapeutics Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
2121
|
Zimmer L, Eigentler TK, Kiecker F, Simon J, Utikal J, Mohr P, Berking C, Kämpgen E, Dippel E, Stadler R, Hauschild A, Fluck M, Terheyden P, Rompel R, Loquai C, Assi Z, Garbe C, Schadendorf D. Open-label, multicenter, single-arm phase II DeCOG-study of ipilimumab in pretreated patients with different subtypes of metastatic melanoma. J Transl Med 2015; 13:351. [PMID: 26541511 PMCID: PMC4635983 DOI: 10.1186/s12967-015-0716-5] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Accepted: 10/26/2015] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Ipilimumab is an approved immunotherapy that has shown an overall survival benefit in patients with cutaneous metastatic melanoma in two phase III trials. As results of registrational trials might not answer all questions regarding safety and efficacy of ipilimumab in patients with advanced melanoma seen in daily clinical practice, the Dermatologic Cooperative Oncology Group conducted a phase II study to assess the efficacy and safety of ipilimumab in patients with different subtypes of metastatic melanoma. PATIENTS AND METHODS We undertook a multicenter phase II study in melanoma patients irrespective of location of the primary melanoma. Here we present data on patients with pretreated metastatic cutaneous, mucosal and occult melanoma 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 103 pretreated patients received at least one dose of ipilimumab, including 83 cutaneous, seven mucosal and 13 occult melanomas. 1-year OS rates for cutaneous, mucosal and occult melanoma were 38 %, 14 % and 27 %, respectively. Median OS was 6.8 months (95 % CI 5.3-9.9) for cutaneous, 9.6 months (95 % CI 1.6-11.1) for mucosal, and 9.9 months (lower 95 % CI 2.3, upper 95 % CI non-existent) for occult melanoma. Overall response rates for cutaneous, mucosal and occult melanoma were 16 %, 17 % and 11 %, respectively. Eleven patients had partial response (16 %) and ten patients experienced stable disease (14 %), none achieved a complete response. Treatment-related AEs were observed in 71 patients (69 %), including 20 grade 3-4 events (19 %). No new and unexpected safety findings were noted. CONCLUSIONS Ipilimumab is a treatment option for pretreated patients with advanced cutaneous melanoma seen in daily routine. Toxicity was manageable when treated as per protocol-specific guidelines. TRIAL REGISTRATION Clinical Trials.gov NCT01355120.
Collapse
Affiliation(s)
- Lisa Zimmer
- Department of Dermatology, University Hospital, University Duisburg-Essen, Essen, Germany.
| | - Thomas K Eigentler
- Department of Dermatology, Center for Dermatooncology, University Medical Center Tübingen, Tübingen, Germany.
| | - Felix Kiecker
- Department of Dermatology and Allergy, Skin Cancer Center, Charité-Universitätsmedizin Berlin, Berlin, Germany.
| | - Jan Simon
- Department of Dermatology, Venereology and Allergology, University Hospital Leipzig, Leipzig, Germany.
| | - Jochen Utikal
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany.
| | - Peter Mohr
- Department of Dermatology, Elbekliniken Stade Buxtehude, Buxtehude, Germany.
| | - Carola Berking
- Department of Dermatology and Allergy, Ludwig-Maximilian University, Munich, Germany.
| | - Eckhart Kämpgen
- Department of Dermatology, Dermatologikum Berlin, Berlin, Germany.
| | - Edgar Dippel
- Department of Dermatology, Klinikum Ludwigshafen, Skin Cancer Center Rheinpfalz, Ludwigshafen, Germany.
| | - Rudolf Stadler
- Department of Dermatology, Medical Centre Minden, Minden, Germany.
| | | | - Michael Fluck
- Department of Dermatology Hornheide, Münster, Germany.
| | | | - Rainer Rompel
- Department of Dermatology, Clinical Centre Kassel, Kassel, Germany.
| | - Carmen Loquai
- Department of Dermatology, University Medical Center, Johannes Gutenberg-University, Mainz, Germany.
| | - Zeinab Assi
- Department of Dermatology, Center for Dermatooncology, University Medical Center Tübingen, Tübingen, Germany.
| | - Claus Garbe
- Department of Dermatology, Center for Dermatooncology, University Medical Center Tübingen, Tübingen, Germany.
| | - Dirk Schadendorf
- Department of Dermatology, University Hospital, University Duisburg-Essen, Essen, Germany.
| |
Collapse
|
2122
|
Sun Z, Zhu Y, Xia J, Sawakami T, Kokudo N, Zhang N. Status of and prospects for cancer vaccines against hepatocellular carcinoma in clinical trials. Biosci Trends 2015; 10:85-91. [PMID: 26522694 DOI: 10.5582/bst.2015.01128] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Current therapies to treat advanced hepatocellular carcinoma (HCC) are not satisfactory because of the high rate of recurrence after treatment and because of severe complications after surgery. Cancer vaccines have been studied for decades to achieve effective, micro-invasive, long-lasting anti-tumor action. Cancer vaccines are designed to promote tumor-specific immune responses and increase specific cytotoxic CD8-positive T cells. This review summarizes 16 phase I clinical trials of cancer vaccines against HCC that have been conducted over the past 10 years. According to those trials, the Alpha fetoprotein (AFP), Glypican-3 (GPC3), and Multidrug resistance-associated protein 3 (MRP3) vaccines were well tolerated and safe. Some early clinical trials have shown that vaccination resulted in a large number of T cells activated by a specific tumor-associated antigen in the circulation, but clinical outcomes were not satisfactory. This may be because targets for immunosuppressive agents have yet to be clearly determined in HCC. Therapeutic regimens that combine activative agents and suppressive agents may profoundly improve clinical outcomes for patients with HCC in the future.
Collapse
Affiliation(s)
- Zhipeng Sun
- Beijing Key Lab of Therapeutic Cancer Vaccines, Peking University Ninth School of Clinical Medicine (Cancer Center, BeijingShijitan Hospital, Capital Medical University)
| | | | | | | | | | | |
Collapse
|
2123
|
Kurtulus S, Sakuishi K, Ngiow SF, Joller N, Tan DJ, Teng MW, Smyth MJ, Kuchroo VK, Anderson AC. TIGIT predominantly regulates the immune response via regulatory T cells. J Clin Invest 2015; 125:4053-62. [PMID: 26413872 PMCID: PMC4639980 DOI: 10.1172/jci81187] [Citation(s) in RCA: 433] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 08/17/2015] [Indexed: 12/22/2022] Open
Abstract
Coinhibitory receptors are critical for the maintenance of immune homeostasis. Upregulation of these receptors on effector T cells terminates T cell responses, while their expression on Tregs promotes their suppressor function. Understanding the function of coinhibitory receptors in effector T cells and Tregs is crucial, as therapies that target coinhibitory receptors are currently at the forefront of treatment strategies for cancer and other chronic diseases. T cell Ig and ITIM domain (TIGIT) is a recently identified coinhibitory receptor that is found on the surface of a variety of lymphoid cells, and its role in immune regulation is just beginning to be elucidated. We examined TIGIT-mediated immune regulation in different murine cancer models and determined that TIGIT marks the most dysfunctional subset of CD8+ T cells in tumor tissue as well as tumor-tissue Tregs with a highly active and suppressive phenotype. We demonstrated that TIGIT signaling in Tregs directs their phenotype and that TIGIT primarily suppresses antitumor immunity via Tregs and not CD8+ T cells. Moreover, TIGIT+ Tregs upregulated expression of the coinhibitory receptor TIM-3 in tumor tissue, and TIM-3 and TIGIT synergized to suppress antitumor immune responses. Our findings provide mechanistic insight into how TIGIT regulates immune responses in chronic disease settings.
Collapse
MESH Headings
- Adoptive Transfer
- Animals
- Colonic Neoplasms/immunology
- Colonic Neoplasms/pathology
- DNA-Binding Proteins/deficiency
- Female
- Gene Expression Regulation, Neoplastic
- Hepatitis A Virus Cellular Receptor 2
- Immunophenotyping
- Lymphocytes, Tumor-Infiltrating/immunology
- Melanoma, Experimental/immunology
- Melanoma, Experimental/pathology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- Neoplasm Proteins/biosynthesis
- Neoplasm Proteins/genetics
- Receptors, Chemokine/biosynthesis
- Receptors, Chemokine/genetics
- Receptors, Immunologic/deficiency
- Receptors, Immunologic/physiology
- Receptors, Virus/biosynthesis
- Receptors, Virus/genetics
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Regulatory/immunology
- Transcription Factors/biosynthesis
- Transcription Factors/genetics
Collapse
Affiliation(s)
- Sema Kurtulus
- Evergrande Center for Immunologic Diseases and Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Kaori Sakuishi
- Evergrande Center for Immunologic Diseases and Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Shin-Foong Ngiow
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
- School of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Nicole Joller
- Evergrande Center for Immunologic Diseases and Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Dewar J. Tan
- Evergrande Center for Immunologic Diseases and Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Michele W.L. Teng
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
- School of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Mark J. Smyth
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
- School of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Vijay K. Kuchroo
- Evergrande Center for Immunologic Diseases and Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Ana C. Anderson
- Evergrande Center for Immunologic Diseases and Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
2124
|
Fakhrejahani F, Tomita Y, Maj-Hes A, Trepel JB, De Santis M, Apolo AB. Immunotherapies for bladder cancer: a new hope. Curr Opin Urol 2015; 25:586-96. [PMID: 26372038 PMCID: PMC6777558 DOI: 10.1097/mou.0000000000000213] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW We review recent data on immunotherapies for bladder cancer and discuss strategies to maximize the antitumor effect of immunotherapy in solid tumors. RECENT FINDINGS Anti-programmed death ligand 1 has shown promise in advanced bladder cancer. Clinical trials of immune checkpoint inhibitors as monotherapy or in combination are underway. Here we review strategies for enhancing antitumor immunity using immunomodulating agents or combination treatments that may increase tumor response. SUMMARY Combining immune checkpoint inhibitors with other treatment modalities may lead to the development of new treatment strategies in advanced bladder cancer; however, identifying predictive biomarkers is essential for appropriate patient selection.
Collapse
Affiliation(s)
- Farhad Fakhrejahani
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Yusuke Tomita
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Agnes Maj-Hes
- Department of Urology, Medical University of Vienna, Vienna, Austria
| | - Jane B. Trepel
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Maria De Santis
- Cancer Research Unit, Warwick University Medical School, Coventry, UK
| | - Andrea B. Apolo
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
2125
|
Cherington CC, Bryce AH. The end of the beginning: PD-1 inhibition as the new standard of care first-line immunotherapy in metastatic melanoma. Melanoma Manag 2015; 2:305-309. [PMID: 30190858 PMCID: PMC6094642 DOI: 10.2217/mmt.15.32] [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: 01/19/2023] Open
Affiliation(s)
- Chad C Cherington
- Department of Medical Oncology, Mayo Clinic, 13400 East Shea Boulevard, Scottsdale, AZ 85259, USA
| | - Alan H Bryce
- Department of Medical Oncology, Mayo Clinic, 13400 East Shea Boulevard, Scottsdale, AZ 85259, USA
| |
Collapse
|
2126
|
Munhoz RR, González AF, Reed VA, Postow MA. Targeting immune checkpoints in melanoma: an update. Melanoma Manag 2015; 2:339-352. [PMID: 30190862 PMCID: PMC6094685 DOI: 10.2217/mmt.15.25] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Different treatment modalities encompassed under the term 'immunotherapy' have led to major breakthroughs in the treatment of melanoma. Immune checkpoint-blocking antibodies targeting CTLA-4 and PD-1 result in significant activity and prolonged survival in patients with advanced melanoma and are currently available for clinical use. Studies addressing novel immune checkpoint blocking antibodies, combined approaches and predictive/prognostic biomarkers are expected to broaden the applicability and efficacy of this approach. In this article, we will review clinically meaningful aspects of immune checkpoint blockade, promising strategies under development and the challenges faced in a continuous search to improve the outcomes of patients affected by this disease.
Collapse
Affiliation(s)
- Rodrigo R Munhoz
- Melanoma & Immunotherapeutics Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Instituto do Câncer do Estado de São Paulo, Universidade de São Paulo, Brazil
| | | | - Vanessa A Reed
- Melanoma & Immunotherapeutics Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Michael A Postow
- Melanoma & Immunotherapeutics Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
2127
|
Yamazaki N, Uhara H, Fukushima S, Uchi H, Shibagaki N, Kiyohara Y, Tsutsumida A, Namikawa K, Okuyama R, Otsuka Y, Tokudome T. Phase II study of the immune-checkpoint inhibitor ipilimumab plus dacarbazine in Japanese patients with previously untreated, unresectable or metastatic melanoma. Cancer Chemother Pharmacol 2015; 76:969-75. [PMID: 26407818 PMCID: PMC4612320 DOI: 10.1007/s00280-015-2870-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 09/06/2015] [Indexed: 11/24/2022]
Abstract
PURPOSE Ipilimumab (IPI), a monoclonal antibody against immune-checkpoint receptor cytotoxic T lymphocyte antigen-4, is designed to enhance antitumor T cell function. IPI 10 mg/kg plus dacarbazine (DTIC) significantly improved overall survival in a phase 3 study involving predominantly Caucasian patients, with an adverse event (AE) profile similar to that of IPI monotherapy. We conducted a single-arm, phase 2 study to evaluate the safety and efficacy of IPI plus DTIC in Japanese patients. METHODS Previously untreated patients with unresectable stage III or IV melanoma received IPI 10 mg/kg plus DTIC 850 mg/m(2) every 3 weeks for four doses (q3w × 4), followed by DTIC q3w × 4 and then IPI every 12 weeks until disease progression or intolerable toxicity. RESULTS All 15 treated patients reported drug-related AEs, the most common of which were increases in alanine aminotransferase (n = 12, 80 %) and aspartate aminotransferase (n = 11, 73 %). Treatment-related serious AEs were reported in 11 (73 %) patients. Nine patients (60 %) discontinued treatment due to drug-related toxicities. Immune-related AEs (irAEs) were reported in 14 patients (93 %). The most frequent irAEs were liver (n = 12, 80 %) and skin (n = 10, 67 %) toxicities. Five deaths were reported; all were caused by progressive disease. Efficacy evaluation showed one complete response, one partial response and four patients with stable disease. Best overall response rate was 13 % (2/15), and the disease control rate was 40 % (6/15). The study was terminated early due to frequent, high-grade liver toxicities. CONCLUSIONS IPI 10 mg/kg plus DTIC 850 mg/m(2) was not considered tolerable in the Japanese patient population. ClinicalTrials.gov identifier: NCT01681212.
Collapse
Affiliation(s)
- N Yamazaki
- Department of Dermatologic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - H Uhara
- Department of Dermatology, Shinshu University School of Medicine, Matsumoto, Japan
| | - S Fukushima
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - H Uchi
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - N Shibagaki
- Department of Dermatology, Yamanashi University Hospital, Yamanashi, Japan
| | - Y Kiyohara
- Dermatology Division, Shizuoka Cancer Center, Shizuoka, Japan
| | - A Tsutsumida
- Department of Dermatologic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - K Namikawa
- Department of Dermatologic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - R Okuyama
- Department of Dermatology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Y Otsuka
- Research and Development, Bristol-Myers K.K., 6-5-1, Nishishinjuku, Shinjuku-ku, Tokyo, 163-1328, Japan
| | - T Tokudome
- Research and Development, Bristol-Myers K.K., 6-5-1, Nishishinjuku, Shinjuku-ku, Tokyo, 163-1328, Japan.
| |
Collapse
|
2128
|
Tsai KK, Daud AI. Nivolumab plus ipilimumab in the treatment of advanced melanoma. J Hematol Oncol 2015; 8:123. [PMID: 26518223 PMCID: PMC4628394 DOI: 10.1186/s13045-015-0219-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 10/13/2015] [Indexed: 12/17/2022] Open
Abstract
Advanced melanoma has historically been a difficult disease to treat due to few effective systemic treatment options. However, over the past few years, scientific advancements in immune checkpoint inhibition have resulted in several novel approaches that have changed front-line management of advanced melanoma. Despite these exciting developments, there remains room for improvement in treatment outcomes. Combination immunotherapy, in particular combined cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and programmed death 1 (PD-1) blockade, represents an important first step in this direction.
Collapse
Affiliation(s)
- Katy K Tsai
- Helen Diller Comprehensive Cancer Center, University of California, 1600 Divisadero St., Box 1770, San Francisco, CA, 94115, USA.
| | - Adil I Daud
- Helen Diller Comprehensive Cancer Center, University of California, 1600 Divisadero St., Box 1770, San Francisco, CA, 94115, USA.
| |
Collapse
|
2129
|
Geukes Foppen MH, Donia M, Svane IM, Haanen JBAG. Tumor-infiltrating lymphocytes for the treatment of metastatic cancer. Mol Oncol 2015; 9:1918-35. [PMID: 26578452 DOI: 10.1016/j.molonc.2015.10.018] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 10/20/2015] [Indexed: 12/15/2022] Open
Abstract
Over the past few years melanoma incidence has been rising steadily, resulting in an increase in melanoma related mortality. Until recently, therapeutic options for metastatic melanoma were scarce. Chemotherapy and, in some countries, IL-2 were the only registered treatment modalities. In the last five years, treatment with immunotherapy (anti CTLA-4, anti PD-1, or the combination of these antibodies) has shown very promising results and was able to improve survival in patients with metastatic melanoma. Adoptive cell therapy using tumor-infiltrating lymphocytes is yet another, but highly promising, immunotherapeutic strategy for patients with metastatic melanoma. This review will discuss the development of TIL as a treatment option for melanoma, its mode of action and simplification over time, and the possibilities to expand this therapy to other types of cancer. Also, the future directions of TIL based therapies will be highlighted.
Collapse
Affiliation(s)
- M H Geukes Foppen
- Department of Medical Oncology, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands.
| | - M Donia
- Center for Cancer Immune Therapy, Department of Haematology and Oncology, Herlev Hospital, University of Copenhagen, Herlev Ringvej 75, 2730 Herlev, Denmark.
| | - I M Svane
- Center for Cancer Immune Therapy, Department of Haematology and Oncology, Herlev Hospital, University of Copenhagen, Herlev Ringvej 75, 2730 Herlev, Denmark.
| | - J B A G Haanen
- Department of Medical Oncology, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands.
| |
Collapse
|
2130
|
The use of combinations of monoclonal antibodies in clinical oncology. Cancer Treat Rev 2015; 41:859-67. [PMID: 26547132 DOI: 10.1016/j.ctrv.2015.10.008] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Revised: 10/21/2015] [Accepted: 10/23/2015] [Indexed: 01/18/2023]
Abstract
Treatment with monoclonal antibodies is becoming increasingly important in clinical oncology. These antibodies specifically inhibit signaling pathways in tumor growth and/or induce immunological responses against tumor cells. By combining monoclonal antibodies several pathways may be targeted simultaneously, potentially leading to additive or synergistic effects. Theoretically, antibodies are very suitable for use in combination therapy, because of limited overlapping toxicity and lack of pharmacokinetic interactions. In this article an overview is given of preclinical and clinical data on twenty-five different combinations of antibodies in oncology. Some of these combinations have proven clinical benefit, for example the combination of trastuzumab and pertuzumab in HER2-positive breast cancer, which exemplifies an additive or synergistic effect on antitumor activity in clinical studies and the combination of nivolumab and ipilimumab, which results in significant increases in progression-free and overall survival in patients with advanced melanoma. However, other combinations may lead to unfavorable results, such as bevacizumab with cetuximab or panitumumab in advanced colorectal cancer. These combinations result in shorter progression-free survival and increased toxicity compared to therapy with a single antibody. In summary, the different published studies showed widely varying results, depending on the combination of antibodies, indication and patient population. More preclinical and clinical studies are necessary to unravel the mechanisms behind synergistic or antagonistic effects of combining monoclonal antibodies. Most research on combination therapies is still in an early stage, but it is expected that for several tumor types the use of combination therapy of antibodies will become standard of care in the near future.
Collapse
|
2131
|
Abdel-Rahman O, ElHalawani H, Fouad M. Risk of gastrointestinal complications in cancer patients treated with immune checkpoint inhibitors: a meta-analysis. Immunotherapy 2015; 7:1213-27. [PMID: 26513491 DOI: 10.2217/imt.15.87] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
AIM We performed a meta-analysis of the risk of selected gastrointestinal toxicities associated with immune checkpoint inhibitors. PATIENTS & METHODS Eligible studies included randomized trials of patients with solid tumors on ipilimumab, nivolumab, pembrolizumab, tremelimumab, pidilizumab and atezolizumab, describing events of diarrhea, vomiting or colitis. RESULTS After exclusion of ineligible studies, a total of ten clinical trials were considered eligible for the meta-analysis. The relative risk of all-grade diarrhea, vomiting and colitis was 1.64 (95% CI: 1.19-2.26; p = 0.002), 0.72 (95% CI: 0.49-1.07; p = 0.1), 10.35 (95% CI: 5.78-18.53; p < 0.00001), respectively. CONCLUSION Our meta-analysis has demonstrated that immune checkpoint inhibitors are associated with a significantly increased risk of all grade and high-grade colitis.
Collapse
Affiliation(s)
- Omar Abdel-Rahman
- Clinical Oncology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Hesham ElHalawani
- Clinical Oncology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Mona Fouad
- Medical Microbiology & Immunology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| |
Collapse
|
2132
|
Tran E, Ahmadzadeh M, Lu YC, Gros A, Turcotte S, Robbins PF, Gartner JJ, Zheng Z, Li YF, Ray S, Wunderlich JR, Somerville RP, Rosenberg SA. Immunogenicity of somatic mutations in human gastrointestinal cancers. Science 2015; 350:1387-90. [PMID: 26516200 DOI: 10.1126/science.aad1253] [Citation(s) in RCA: 593] [Impact Index Per Article: 59.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 10/15/2015] [Indexed: 12/11/2022]
Abstract
It is unknown whether the human immune system frequently mounts a T cell response against mutations expressed by common epithelial cancers. Using a next-generation sequencing approach combined with high-throughput immunologic screening, we demonstrated that tumor-infiltrating lymphocytes (TILs) from 9 out of 10 patients with metastatic gastrointestinal cancers contained CD4(+) and/or CD8(+) T cells that recognized one to three neo-epitopes derived from somatic mutations expressed by the patient's own tumor. There were no immunogenic epitopes shared between these patients. However, we identified in one patient a human leukocyte antigen-C*08:02-restricted T cell receptor from CD8(+) TILs that targeted the KRAS(G12D) hotspot driver mutation found in many human cancers. Thus, a high frequency of patients with common gastrointestinal cancers harbor immunogenic mutations that can potentially be exploited for the development of highly personalized immunotherapies.
Collapse
Affiliation(s)
- Eric Tran
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Mojgan Ahmadzadeh
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yong-Chen Lu
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Alena Gros
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Simon Turcotte
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Paul F Robbins
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jared J Gartner
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Zhili Zheng
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yong F Li
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Satyajit Ray
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - John R Wunderlich
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Robert P Somerville
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Steven A Rosenberg
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
2133
|
Galluzzi L, Eggermont A, Kroemer G. Doubling the blockade for melanoma immunotherapy. Oncoimmunology 2015; 5:e1106127. [PMID: 26942094 DOI: 10.1080/2162402x.2015.1106127] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 10/07/2015] [Indexed: 12/19/2022] Open
Affiliation(s)
- 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 Comprehensive Cancer Institute, Villejuif, France
| | | | - 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 Comprehensive Cancer Institute, Villejuif, France; Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France; Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
2134
|
Desrichard A, Snyder A, Chan TA. Cancer Neoantigens and Applications for Immunotherapy. Clin Cancer Res 2015; 22:807-12. [PMID: 26515495 DOI: 10.1158/1078-0432.ccr-14-3175] [Citation(s) in RCA: 165] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 09/18/2015] [Indexed: 12/12/2022]
Abstract
Recent advances in immune checkpoint blockade therapy have revolutionized the treatment of cancer. Tumor-specific antigens that are generated by somatic mutation, neoantigens, can influence patient response to immunotherapy and contribute to tumor shrinkage. Recent evidence demonstrating the success of checkpoint blockade immunotherapy in boosting T-cell reactivity against patient-specific neoantigens constitutes a strong rationale for the development of personalized vaccines against these nonself peptides. With the decreasing cost of next-generation sequencing, peptide manufacturing, and improvement of in silico prediction of peptide immunogenicity, it is increasingly important to evaluate the potential use of neoantigens in both diagnosis and treatment. Specifically, these neoantigens could be useful both as predictors of immune checkpoint blockade therapy response and/or incorporated in therapeutic vaccination strategies.
Collapse
Affiliation(s)
- Alexis Desrichard
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Alexandra Snyder
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York. Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Timothy A Chan
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York. Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York.
| |
Collapse
|
2135
|
Atkins M. Immunotherapy Combinations With Checkpoint Inhibitors in Metastatic Melanoma: Current Approaches and Future Directions. Semin Oncol 2015; 42 Suppl 3:S12-9. [PMID: 26598055 DOI: 10.1053/j.seminoncol.2015.10.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Based on the complexity of the immune response to cancer and the mechanisms of tumor evasion, it is likely that therapeutic modulation of multiple immune-mediated pathways will be needed to maximally induce tumor regression in patients with advanced melanoma. The rationale of using combination checkpoint inhibitor-based regimens may include the concomitant effects on re-activation of T cells, increased trafficking of tumor reactive lymphocytes into the tumor tissue, and enhanced killing of cancer cells. The administration of nivolumab in combination with ipilimumab demonstrated increased response rates, tumor shrinkage, and median progression-free survival using combined therapy compared with either treatment alone. Although toxicity was also increased, this trial established proof of principle that combination immunotherapy could enhance the efficacy seen with single-agent programmed cell death protein-1 (PD-1) pathway blockade for the unselected patient. Current and future trials are evaluating alternative schedules and other combinations of immunotherapies to determine if they could provide similar efficacy with less toxicity. In addition, efforts are underway to determine how best to integrate combination immunotherapy with other treatment approaches for patients with advanced melanoma.
Collapse
Affiliation(s)
- Michael Atkins
- Deputy Director, Georgetown-Lombardi Comprehensive Cancer Center, Professor of Oncology and Medicine (Hematology/Oncology), Georgetown University School of Medicine, Washington, DC.
| |
Collapse
|
2136
|
Mahoney KM, Rennert PD, Freeman GJ. Combination cancer immunotherapy and new immunomodulatory targets. Nat Rev Drug Discov 2015; 14:561-84. [PMID: 26228759 DOI: 10.1038/nrd4591] [Citation(s) in RCA: 995] [Impact Index Per Article: 99.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Targeting immune checkpoints such as programmed cell death protein 1 (PD1), programmed cell death 1 ligand 1 (PDL1) and cytotoxic T lymphocyte antigen 4 (CTLA4) has achieved noteworthy benefit in multiple cancers by blocking immunoinhibitory signals and enabling patients to produce an effective antitumour response. Inhibitors of CTLA4, PD1 or PDL1 administered as single agents have resulted in durable tumour regression in some patients, and combinations of PD1 and CTLA4 inhibitors may enhance antitumour benefit. Numerous additional immunomodulatory pathways as well as inhibitory factors expressed or secreted by myeloid and stromal cells in the tumour microenvironment are potential targets for synergizing with immune checkpoint blockade. Given the breadth of potential targets in the immune system, critical questions to address include which combinations should move forward in development and which patients will benefit from these treatments. This Review discusses the leading drug targets that are expressed on tumour cells and in the tumour microenvironment that allow enhancement of the antitumour immune response.
Collapse
Affiliation(s)
- Kathleen M Mahoney
- 1] Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02215, USA. [2] Division of Haematology and Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, USA. [3]
| | - Paul D Rennert
- 1] SugarCone Biotech, Holliston, Massachusetts 01746, USA. [2] Videre Biotherapeutics, Watertown, Massachusetts 02472, USA. [3]
| | - Gordon J Freeman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02215, USA
| |
Collapse
|
2137
|
Teixidó C, González-Cao M, Karachaliou N, Rosell R. Predictive factors for immunotherapy in melanoma. ANNALS OF TRANSLATIONAL MEDICINE 2015; 3:208. [PMID: 26488004 DOI: 10.3978/j.issn.2305-5839.2015.05.07] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Immunotherapy has emerged as an exciting strategy for cancer treatment. Therapeutic blockade of immune checkpoint regulators favors the ability of T cell responses to increase anti-tumor immunity. The cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) and programmed cell death-1 (PD-1) are two T cell-inhibitory receptors with independent mechanisms of action. Immune checkpoint inhibitors targeting either CTLA-4, PD-1 or its ligand PD-L1 are currently yielding promising results in terms of efficacy in several clinical studies with melanoma patients and are being developed and tested as immunotherapy agents for multiple cancer types. To date, no reliable predictors of activity and efficacy of immunotherapy have yet been identified or validated. Even so, determining which patients derive clinical benefit from immune checkpoint agents remains an important clinical question and efforts to identify predictive markers of response are ongoing. This article reviews the current potential predictive factors for CTLA-4 and PD-1/PD-L1 immune checkpoints inhibitors in melanoma.
Collapse
Affiliation(s)
- Cristina Teixidó
- 1 Pangaea Biotech, 2 Dr Rosell Oncology Institute, Quirón Dexeus University Hospital, 08028 Barcelona, Spain ; 3 Cancer Biology and Precision Medicine Program, Catalan Institute of Oncology, Hospital Germans Trias i Pujol, 08916 Badalona, Spain ; 4 Molecular Oncology Research Foundation (MORe), Barcelona, Spain
| | - Maria González-Cao
- 1 Pangaea Biotech, 2 Dr Rosell Oncology Institute, Quirón Dexeus University Hospital, 08028 Barcelona, Spain ; 3 Cancer Biology and Precision Medicine Program, Catalan Institute of Oncology, Hospital Germans Trias i Pujol, 08916 Badalona, Spain ; 4 Molecular Oncology Research Foundation (MORe), Barcelona, Spain
| | - Niki Karachaliou
- 1 Pangaea Biotech, 2 Dr Rosell Oncology Institute, Quirón Dexeus University Hospital, 08028 Barcelona, Spain ; 3 Cancer Biology and Precision Medicine Program, Catalan Institute of Oncology, Hospital Germans Trias i Pujol, 08916 Badalona, Spain ; 4 Molecular Oncology Research Foundation (MORe), Barcelona, Spain
| | - Rafael Rosell
- 1 Pangaea Biotech, 2 Dr Rosell Oncology Institute, Quirón Dexeus University Hospital, 08028 Barcelona, Spain ; 3 Cancer Biology and Precision Medicine Program, Catalan Institute of Oncology, Hospital Germans Trias i Pujol, 08916 Badalona, Spain ; 4 Molecular Oncology Research Foundation (MORe), Barcelona, Spain
| |
Collapse
|
2138
|
Karpanen T, Olweus J. T-cell receptor gene therapy--ready to go viral? Mol Oncol 2015; 9:2019-42. [PMID: 26548533 DOI: 10.1016/j.molonc.2015.10.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 10/08/2015] [Accepted: 10/09/2015] [Indexed: 12/16/2022] Open
Abstract
T lymphocytes can be redirected to recognize a tumor target and harnessed to combat cancer by genetic introduction of T-cell receptors of a defined specificity. This approach has recently mediated encouraging clinical responses in patients with cancers previously regarded as incurable. However, despite the great promise, T-cell receptor gene therapy still faces a multitude of obstacles. Identification of epitopes that enable effective targeting of all the cells in a heterogeneous tumor while sparing normal tissues remains perhaps the most demanding challenge. Experience from clinical trials has revealed the dangers associated with T-cell receptor gene therapy and highlighted the need for reliable preclinical methods to identify potentially hazardous recognition of both intended and unintended epitopes in healthy tissues. Procedures for manufacturing large and highly potent T-cell populations can be optimized to enhance their antitumor efficacy. Here, we review the current knowledge gained from preclinical models and clinical trials using adoptive transfer of T-cell receptor-engineered T lymphocytes, discuss the major challenges involved and highlight potential strategies to increase the safety and efficacy to make T-cell receptor gene therapy a standard-of-care for large patient groups.
Collapse
Affiliation(s)
- Terhi Karpanen
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet and K.G. Jebsen Center for Cancer Immunotherapy, University of Oslo, Ullernchausseen 70, N-0379 Oslo, Norway.
| | - Johanna Olweus
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet and K.G. Jebsen Center for Cancer Immunotherapy, University of Oslo, Ullernchausseen 70, N-0379 Oslo, Norway.
| |
Collapse
|
2139
|
Van Allen EM, Miao D, Schilling B, Shukla SA, Blank C, Zimmer L, Sucker A, Hillen U, Foppen MHG, Goldinger SM, Utikal J, Hassel JC, Weide B, Kaehler KC, Loquai C, Mohr P, Gutzmer R, Dummer R, Gabriel S, Wu CJ, Schadendorf D, Garraway LA. Genomic correlates of response to CTLA-4 blockade in metastatic melanoma. Science 2015; 350:207-211. [PMID: 26359337 PMCID: PMC5054517 DOI: 10.1126/science.aad0095] [Citation(s) in RCA: 2119] [Impact Index Per Article: 211.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 08/27/2015] [Indexed: 12/12/2022]
Abstract
Monoclonal antibodies directed against cytotoxic T lymphocyte-associated antigen-4 (CTLA-4), such as ipilimumab, yield considerable clinical benefit for patients with metastatic melanoma by inhibiting immune checkpoint activity, but clinical predictors of response to these therapies remain incompletely characterized. To investigate the roles of tumor-specific neoantigens and alterations in the tumor microenvironment in the response to ipilimumab, we analyzed whole exomes from pretreatment melanoma tumor biopsies and matching germline tissue samples from 110 patients. For 40 of these patients, we also obtained and analyzed transcriptome data from the pretreatment tumor samples. Overall mutational load, neoantigen load, and expression of cytolytic markers in the immune microenvironment were significantly associated with clinical benefit. However, no recurrent neoantigen peptide sequences predicted responder patient populations. Thus, detailed integrated molecular characterization of large patient cohorts may be needed to identify robust determinants of response and resistance to immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Eliezer M. Van Allen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Center for Cancer Precision Medicine, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Diana Miao
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Bastian Schilling
- Department of Dermatology, University Hospital, University Duisburg—Essen, 45147 Essen, Germany
- German Cancer Consortium (DKTK), 69121 Heidelberg, Germany
| | - Sachet A. Shukla
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Christian Blank
- Department of Medical Oncology, Netherlands Cancer Institute, 1066 CX Amsterdam, Netherlands
| | - Lisa Zimmer
- Department of Dermatology, University Hospital, University Duisburg—Essen, 45147 Essen, Germany
- German Cancer Consortium (DKTK), 69121 Heidelberg, Germany
| | - Antje Sucker
- Department of Dermatology, University Hospital, University Duisburg—Essen, 45147 Essen, Germany
- German Cancer Consortium (DKTK), 69121 Heidelberg, Germany
| | - Uwe Hillen
- Department of Dermatology, University Hospital, University Duisburg—Essen, 45147 Essen, Germany
- German Cancer Consortium (DKTK), 69121 Heidelberg, Germany
| | - Marnix H. Geukes Foppen
- Department of Medical Oncology, Netherlands Cancer Institute, 1066 CX Amsterdam, Netherlands
| | - Simone M. Goldinger
- Department of Dermatology, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Jochen Utikal
- German Cancer Consortium (DKTK), 69121 Heidelberg, Germany
- Skin Cancer Unit, German Cancer Research Center(DKTK), 69121 Heidelberg, Germany
- Department of Dermatology, Venerology, and Allergology, University Medical Center, Ruprecht-Karls University of Heidelberg, 68167 Mannheim, Germany
| | - Jessica C. Hassel
- Department of Dermatology, University Hospital, Ruprecht-Karls University of Heidelberg, 69120 Heidelberg, Germany
| | - Benjamin Weide
- Department of Dermatology, University Hospital Tübingen, 72076 Tübingen, Germany
| | | | - Carmen Loquai
- Department of Dermatology, University Medical Center, 55131 Mainz, Germany
| | - Peter Mohr
- Department of Dermatology, Elbe-Kliniken, 21614 Buxtehude, Germany
| | - Ralf Gutzmer
- Department of Dermatology and Allergy, Skin Cancer Center Hannover, Hannover Medical School, 30625 Hannover, Germany
| | - Reinhard Dummer
- Department of Dermatology, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Stacey Gabriel
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Catherine J. Wu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Dirk Schadendorf
- Department of Dermatology, University Hospital, University Duisburg—Essen, 45147 Essen, Germany
- German Cancer Consortium (DKTK), 69121 Heidelberg, Germany
| | - Levi A. Garraway
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Center for Cancer Precision Medicine, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| |
Collapse
|
2140
|
Van Allen EM, Miao D, Schilling B, Shukla SA, Blank C, Zimmer L, Sucker A, Hillen U, Foppen MHG, Goldinger SM, Utikal J, Hassel JC, Weide B, Kaehler KC, Loquai C, Mohr P, Gutzmer R, Dummer R, Gabriel S, Wu CJ, Schadendorf D, Garraway LA. Genomic correlates of response to CTLA-4 blockade in metastatic melanoma. Science 2015; 350:207-211. [PMID: 26359337 PMCID: PMC5054517 DOI: 10.1126/science.aad0095 10.1126/science.aaf8264] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 08/27/2015] [Indexed: 07/10/2023]
Abstract
Monoclonal antibodies directed against cytotoxic T lymphocyte-associated antigen-4 (CTLA-4), such as ipilimumab, yield considerable clinical benefit for patients with metastatic melanoma by inhibiting immune checkpoint activity, but clinical predictors of response to these therapies remain incompletely characterized. To investigate the roles of tumor-specific neoantigens and alterations in the tumor microenvironment in the response to ipilimumab, we analyzed whole exomes from pretreatment melanoma tumor biopsies and matching germline tissue samples from 110 patients. For 40 of these patients, we also obtained and analyzed transcriptome data from the pretreatment tumor samples. Overall mutational load, neoantigen load, and expression of cytolytic markers in the immune microenvironment were significantly associated with clinical benefit. However, no recurrent neoantigen peptide sequences predicted responder patient populations. Thus, detailed integrated molecular characterization of large patient cohorts may be needed to identify robust determinants of response and resistance to immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Eliezer M. Van Allen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Center for Cancer Precision Medicine, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Diana Miao
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Bastian Schilling
- Department of Dermatology, University Hospital, University Duisburg—Essen, 45147 Essen, Germany
- German Cancer Consortium (DKTK), 69121 Heidelberg, Germany
| | - Sachet A. Shukla
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Christian Blank
- Department of Medical Oncology, Netherlands Cancer Institute, 1066 CX Amsterdam, Netherlands
| | - Lisa Zimmer
- Department of Dermatology, University Hospital, University Duisburg—Essen, 45147 Essen, Germany
- German Cancer Consortium (DKTK), 69121 Heidelberg, Germany
| | - Antje Sucker
- Department of Dermatology, University Hospital, University Duisburg—Essen, 45147 Essen, Germany
- German Cancer Consortium (DKTK), 69121 Heidelberg, Germany
| | - Uwe Hillen
- Department of Dermatology, University Hospital, University Duisburg—Essen, 45147 Essen, Germany
- German Cancer Consortium (DKTK), 69121 Heidelberg, Germany
| | - Marnix H. Geukes Foppen
- Department of Medical Oncology, Netherlands Cancer Institute, 1066 CX Amsterdam, Netherlands
| | - Simone M. Goldinger
- Department of Dermatology, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Jochen Utikal
- German Cancer Consortium (DKTK), 69121 Heidelberg, Germany
- Skin Cancer Unit, German Cancer Research Center(DKTK), 69121 Heidelberg, Germany
- Department of Dermatology, Venerology, and Allergology, University Medical Center, Ruprecht-Karls University of Heidelberg, 68167 Mannheim, Germany
| | - Jessica C. Hassel
- Department of Dermatology, University Hospital, Ruprecht-Karls University of Heidelberg, 69120 Heidelberg, Germany
| | - Benjamin Weide
- Department of Dermatology, University Hospital Tübingen, 72076 Tübingen, Germany
| | | | - Carmen Loquai
- Department of Dermatology, University Medical Center, 55131 Mainz, Germany
| | - Peter Mohr
- Department of Dermatology, Elbe-Kliniken, 21614 Buxtehude, Germany
| | - Ralf Gutzmer
- Department of Dermatology and Allergy, Skin Cancer Center Hannover, Hannover Medical School, 30625 Hannover, Germany
| | - Reinhard Dummer
- Department of Dermatology, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Stacey Gabriel
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Catherine J. Wu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Dirk Schadendorf
- Department of Dermatology, University Hospital, University Duisburg—Essen, 45147 Essen, Germany
- German Cancer Consortium (DKTK), 69121 Heidelberg, Germany
| | - Levi A. Garraway
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Center for Cancer Precision Medicine, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| |
Collapse
|
2141
|
Rivalland G, Loveland B, Mitchell P. Update on Mucin-1 immunotherapy in cancer: a clinical perspective. Expert Opin Biol Ther 2015; 15:1773-87. [PMID: 26453294 DOI: 10.1517/14712598.2015.1088519] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
INTRODUCTION Mucin 1 (MUC1) is particularly well suited as a cancer immunotherapy target due to the elevated protein expression and aberrant forms associated with malignancy. A variety of therapeutic strategies have been explored, including antibodies intended to induce cancer cell destruction, and vaccinations with peptides, tumor extracts, and gene expression systems. AREAS COVERED MUC1 immunotherapeutic strategies have included vaccination with peptide sequences, glycan molecules, viruses, and dendritic cells, monoclonal antibodies and monoclonal antibody conjugates. Here we review the relevant clinical trials in each field of immunotherapy with particular focus on large and recently published trials. EXPERT OPINION Long clinical experience in the trial setting has reduced concerns of immunotherapy associated toxicities and inappropriate immune responses, with the main limitation (common to many experimental approaches) being a lack of clinical efficacy. However, there have been sufficient treatment-associated responses to justify continued pursuit of MUC1 targeted immunotherapies. The focus now should be on application to the relevant cancers under appropriate circumstances and combination with the emerging non-specific immunotherapy approaches such as the PD-1 pathway inhibitors.
Collapse
Affiliation(s)
- Gareth Rivalland
- a 1 Austin Health, Olivia Newton-John Cancer and Wellness Centre , Studley Rd, Heidelberg VIC 3084, Australia
| | - Bruce Loveland
- b 2 Burnet Institute, Centre for Biomedical Research , Melbourne VIC 3004, Australia
| | - Paul Mitchell
- c 3 Austin Health, Level 4, Olivia Newton-John Cancer and Wellness Centre , Studley Rd, Heidelberg VIC 3084, Australia +613 94 96 57 63 ; +613 94 57 66 98 ;
| |
Collapse
|
2142
|
Freeman-Keller M, Kim Y, Cronin H, Richards A, Gibney G, Weber JS. Nivolumab in Resected and Unresectable Metastatic Melanoma: Characteristics of Immune-Related Adverse Events and Association with Outcomes. Clin Cancer Res 2015; 22:886-94. [PMID: 26446948 DOI: 10.1158/1078-0432.ccr-15-1136] [Citation(s) in RCA: 658] [Impact Index Per Article: 65.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 09/28/2015] [Indexed: 12/17/2022]
Abstract
PURPOSE Retrospective analysis of irAEs in melanoma patients treated with nivolumab. EXPERIMENTAL DESIGN Data were pooled from 148 patients (33 resected, 115 unresectable) treated with nivolumab plus peptide vaccine or nivolumab alone every 2 weeks for 12 weeks. Patients with stable disease or regression received an additional 12-week cycle, then nivolumab alone every 12 weeks for up to 2 additional years. Frequency, grade, and characteristics of immune-related adverse events (irAE) were analyzed. A 12-week landmark survival analysis using a multivariate time-dependent Cox proportional hazard model assessed difference in overall survival (OS) in the presence or absence of irAEs. RESULTS IrAEs of any grade were observed in 68.2% of patients (101 of 148). Grade III/IV irAEs were infrequent: 3 (2%) had grade III rash, 2 (1.35%) had asymptomatic grade III elevation in amylase/lipase, and 2 (1.35%) had grade III colitis. A statistically significant OS difference was noted among patients with any grade of irAE versus those without (P ≤ 0.001), and OS benefit was noted in patients who reported three or more irAE events (P ≤ 0.001). Subset analyses showed statistically significant OS differences with rash [P = 0.001; HR, 0.423; 95% confidence interval (CI), 0.243-0.735] and vitiligo (P = 0.012; HR, 0.184; 95% CI, 0.036-0.94). Rash and vitiligo also correlated with statistically significant OS differences in patients with metastatic disease (P = 0.004 and P = 0.028, respectively). No significant survival differences were seen with other irAEs (endocrinopathies, colitis, or pneumonitis). CONCLUSIONS Cutaneous irAEs are associated with improved survival in melanoma patients treated with nivolumab, and clinical benefit should be validated in larger prospective analyses.
Collapse
Affiliation(s)
| | - Youngchul Kim
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, Florida
| | - Heather Cronin
- Clinical Trials Office, Moffitt Cancer Center, Tampa, Florida
| | | | - Geoffrey Gibney
- Department of Cutaneous Oncology, Lombardi Comprehensive Cancer Center, Washington, DC
| | - Jeffrey S Weber
- Donald A. Adam Comprehensive Melanoma Research Center, Moffitt Cancer Center, Tampa, Florida
| |
Collapse
|
2143
|
Kersten K, Salvagno C, de Visser KE. Exploiting the Immunomodulatory Properties of Chemotherapeutic Drugs to Improve the Success of Cancer Immunotherapy. Front Immunol 2015; 6:516. [PMID: 26500653 PMCID: PMC4595807 DOI: 10.3389/fimmu.2015.00516] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 09/22/2015] [Indexed: 12/29/2022] Open
Abstract
Cancer immunotherapy is gaining momentum in the clinic. The current challenge is to understand why a proportion of cancer patients do not respond to cancer immunotherapy, and how this can be translated into the rational design of combinatorial cancer immunotherapy strategies aimed at maximizing success of immunotherapy. Here, we discuss how tumors orchestrate an immunosuppressive microenvironment, which contributes to their escape from immune attack. Relieving the immunosuppressive networks in cancer patients is an attractive strategy to extend the clinical success of cancer immunotherapy. Since the clinical availability of drugs specifically targeting immunosuppressive cells or mediators is still limited, an alternative strategy is to use conventional chemotherapy drugs with immunomodulatory properties to improve cancer immunotherapy. We summarize the preclinical and clinical studies that illustrate how the anti-tumor T cell response can be enhanced by chemotherapy-induced relief of immunosuppressive networks. Treatment strategies aimed at combining chemotherapy-induced relief of immunosuppression and T cell-boosting checkpoint inhibitors provide an attractive and clinically feasible approach to overcome intrinsic and acquired resistance to cancer immunotherapy, and to extend the clinical success of cancer immunotherapy.
Collapse
Affiliation(s)
- Kelly Kersten
- Division of Immunology, Netherlands Cancer Institute , Amsterdam , Netherlands
| | - Camilla Salvagno
- Division of Immunology, Netherlands Cancer Institute , Amsterdam , Netherlands
| | - Karin E de Visser
- Division of Immunology, Netherlands Cancer Institute , Amsterdam , Netherlands
| |
Collapse
|
2144
|
Grob JJ, Long GV, Schadendorf D, Flaherty K. Disease kinetics for decision-making in advanced melanoma: a call for scenario-driven strategy trials. Lancet Oncol 2015; 16:e522-6. [DOI: 10.1016/s1470-2045(15)00003-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Revised: 05/20/2015] [Accepted: 05/21/2015] [Indexed: 11/26/2022]
|
2145
|
Baselga J, Bhardwaj N, Cantley LC, DeMatteo R, DuBois RN, Foti M, Gapstur SM, Hahn WC, Helman LJ, Jensen RA, Paskett ED, Lawrence TS, Lutzker SG, Szabo E. AACR Cancer Progress Report 2015. Clin Cancer Res 2015; 21:S1-128. [PMID: 26429991 PMCID: PMC5001568 DOI: 10.1158/1078-0432.ccr-15-1846] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
2146
|
Rogiers A, van den Oord JJ, Garmyn M, Stas M, Kenis C, Wildiers H, Marine JC, Wolter P. Novel Therapies for Metastatic Melanoma: An Update on Their Use in Older Patients. Drugs Aging 2015; 32:821-34. [PMID: 26442859 DOI: 10.1007/s40266-015-0304-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cutaneous melanoma is the most aggressive form of skin cancer. With age as a risk factor, melanoma is projected to become a substantial healthcare burden. The clinical course of melanoma in older patients is different from that in middle-aged and younger patients: melanomas are thicker, have higher mitotic rates and are more likely to be ulcerated. Older patients also have a higher mortality rate, yet, paradoxically, have a lower rate of lymph node metastases. After decades of no significant progress in the treatment of this devastating disease, novel insights into the mechanisms underlying the pathophysiology of metastatic melanoma have led to new and remarkably efficient therapeutic opportunities. The discovery that about half of all melanomas carry BRAF mutations led to the introduction of targeted therapy with significant improvements in clinical outcomes. Although these drugs appear to be equally effective in older patients, specific considerations regarding adverse events are required. Besides targeted therapy, immunotherapy has emerged as an alternative therapeutic option. Antibodies that block cytotoxic T-lymphocyte antigen 4 (CTLA-4) and programmed cell death protein 1 (PD-1) can induce responses with high durability. Despite an aging immune system, older patients seem to benefit to the same degree from these treatments, apparently without increased toxicity. In this review, we focus on the epidemiology, clinicopathological features, and recent developments of systemic treatment in cutaneous melanoma with regard to older patients.
Collapse
Affiliation(s)
- Aljosja Rogiers
- Department of General Medical Oncology, University Hospitals Leuven, Herestraat 49, 3000, Leuven, Belgium
- Laboratory for Molecular Cancer Biology, Center for the Biology of Disease, VIB, Leuven, Belgium
- Center for Human Genetics, KU Leuven, Leuven, Belgium
| | | | - Marjan Garmyn
- Department of Dermatology, University Hospitals Leuven, Leuven, Belgium
| | - Marguerite Stas
- Department of Surgical Oncology, University Hospitals Leuven, Leuven, Belgium
| | - Cindy Kenis
- Department of General Medical Oncology, University Hospitals Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Hans Wildiers
- Department of General Medical Oncology, University Hospitals Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Jean-Christophe Marine
- Laboratory for Molecular Cancer Biology, Center for the Biology of Disease, VIB, Leuven, Belgium
- Center for Human Genetics, KU Leuven, Leuven, Belgium
| | - Pascal Wolter
- Department of General Medical Oncology, University Hospitals Leuven, Herestraat 49, 3000, Leuven, Belgium.
| |
Collapse
|
2147
|
Karachaliou N, Pilotto S, Teixidó C, Viteri S, González-Cao M, Riso A, Morales-Espinosa D, Molina MA, Chaib I, Santarpia M, Richardet E, Bria E, Rosell R. Melanoma: oncogenic drivers and the immune system. ANNALS OF TRANSLATIONAL MEDICINE 2015; 3:265. [PMID: 26605311 PMCID: PMC4630557 DOI: 10.3978/j.issn.2305-5839.2015.08.06] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 08/04/2015] [Indexed: 12/19/2022]
Abstract
Advances and in-depth understanding of the biology of melanoma over the past 30 years have contributed to a change in the consideration of melanoma as one of the most therapy-resistant malignancies. The finding that oncogenic BRAF mutations drive tumor growth in up to 50% of melanomas led to a molecular therapy revolution for unresectable and metastatic disease. Moving beyond BRAF, inactivation of immune regulatory checkpoints that limit T cell responses to melanoma has provided targets for cancer immunotherapy. In this review, we discuss the molecular biology of melanoma and we focus on the recent advances of molecularly targeted and immunotherapeutic approaches.
Collapse
|
2148
|
Targeting immune checkpoints: New opportunity for mesothelioma treatment? Cancer Treat Rev 2015; 41:914-24. [PMID: 26433514 DOI: 10.1016/j.ctrv.2015.09.006] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Revised: 09/18/2015] [Accepted: 09/22/2015] [Indexed: 02/07/2023]
Abstract
Malignant pleural mesothelioma is an aggressive cancer linked to asbestos exposure in most patients. Due to the long latency between exposure and presentation, incidence is expected to further increase in the next decade, despite the ban on asbestos import which occurred at the end of last century in industrialized countries. Platinum-based palliative chemotherapy is the only treatment with proven benefit on outcome, resulting in selected patients in a median overall survival of about 1 year. Therefore, there is room for therapeutic improvement using a new strategy to prolong survival. Dealing with cancer cell induced immunosuppression is a promising approach. Reactivating immune responses that are silenced by immune checkpoints recently gained a lot of interest. Checkpoint blockade has already shown promising preclinical and clinical results in several cancer types and is currently also being investigated in mesothelioma. Here, we discuss the expression patterns and mechanisms of action of CTLA-4 and PD-1 as the two most studied and of TIM-3 and LAG-3 as two interesting upcoming immune checkpoints. Furthermore, we review the clinical results of molecules blocking these immune checkpoints and point out their future opportunities with a special focus on mesothelioma.
Collapse
|
2149
|
Phase II study of ipilimumab monotherapy in Japanese patients with advanced melanoma. Cancer Chemother Pharmacol 2015; 76:997-1004. [PMID: 26410424 PMCID: PMC4612321 DOI: 10.1007/s00280-015-2873-x] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 09/09/2015] [Indexed: 10/29/2022]
Abstract
PURPOSE Ipilimumab is designed to block cytotoxic T-lymphocyte antigen-4 to augment antitumor T cell responses. In studies of predominantly Caucasian patients with advanced melanoma, ipilimumab was associated with durable response, long-term survival benefit, and a manageable safety profile. This phase II study assessed the safety of ipilimumab in Japanese patients with unresectable stage III or IV melanoma. METHODS Patients received ipilimumab 3 mg/kg every 3 weeks for four doses. The database lock for the original analysis was in August 2014. Overall survival, progression-free survival, and data on deaths were based on an updated, follow-up analysis (database lock April 2015). RESULTS Data are reported from 20 patients. Fifteen patients (75 %) received all four doses of ipilimumab during induction. Twelve patients (60 %) had at least one drug-related adverse event (AE), and no patients discontinued due to a drug-related AE. There were no deaths related to study drug. The most common drug-related AEs were rash (n = 7), pyrexia (n = 3), increased aspartate aminotransferase (AST; n = 3), and increased alanine aminotransferase (ALT; n = 3). Twelve patients (60 %) reported immune-related AEs (irAEs); most frequent were skin (n = 9) and liver (n = 3) disorders. Grade 3 irAEs were ALT and AST elevation (n = 2) and diabetes mellitus (n = 1). Two patients had a partial response and two had stable disease, yielding a 20 % disease control rate. Median overall survival and progression-free survival were 8.71 and 2.74 months, respectively. CONCLUSION Ipilimumab 3 mg/kg had a manageable AE profile in this Japanese patient population with clinical outcomes similar to that in Caucasian patients. CLINICALTRIALS. GOV IDENTIFIER NCT01990859.
Collapse
|
2150
|
Gallagher SJ, Tiffen JC, Hersey P. Histone Modifications, Modifiers and Readers in Melanoma Resistance to Targeted and Immune Therapy. Cancers (Basel) 2015; 7:1959-82. [PMID: 26426052 PMCID: PMC4695870 DOI: 10.3390/cancers7040870] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 09/17/2015] [Accepted: 09/18/2015] [Indexed: 02/06/2023] Open
Abstract
The treatment of melanoma has been revolutionized by new therapies targeting MAPK signaling or the immune system. Unfortunately these therapies are hindered by either primary resistance or the development of acquired resistance. Resistance mechanisms involving somatic mutations in genes associated with resistance have been identified in some cases of melanoma, however, the cause of resistance remains largely unexplained in other cases. The importance of epigenetic factors targeting histones and histone modifiers in driving the behavior of melanoma is only starting to be unraveled and provides significant opportunity to combat the problems of therapy resistance. There is also an increasing ability to target these epigenetic changes with new drugs that inhibit these modifications to either prevent or overcome resistance to both MAPK inhibitors and immunotherapy. This review focuses on changes in histones, histone reader proteins and histone positioning, which can mediate resistance to new therapeutics and that can be targeted for future therapies.
Collapse
Affiliation(s)
- Stuart J Gallagher
- Melanoma Immunology and Oncology Group, Centenary Institute, University of Sydney, Camperdown 2050, Australia.
- Melanoma Institute Australia, Crow's Nest 2065, Sydney, Australia.
| | - Jessamy C Tiffen
- Melanoma Immunology and Oncology Group, Centenary Institute, University of Sydney, Camperdown 2050, Australia.
- Melanoma Institute Australia, Crow's Nest 2065, Sydney, Australia.
| | - Peter Hersey
- Melanoma Immunology and Oncology Group, Centenary Institute, University of Sydney, Camperdown 2050, Australia.
- Melanoma Institute Australia, Crow's Nest 2065, Sydney, Australia.
| |
Collapse
|