201
|
Queirolo P, Spagnolo F. BRAF plus MEK-targeted drugs: a new standard of treatment for BRAF-mutant advanced melanoma. Cancer Metastasis Rev 2017; 36:35-42. [DOI: 10.1007/s10555-017-9660-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
202
|
Letendre P, Monga V, Milhem M, Zakharia Y. Ipilimumab: from preclinical development to future clinical perspectives in melanoma. Future Oncol 2017; 13:625-636. [PMID: 27882779 PMCID: PMC5618953 DOI: 10.2217/fon-2016-0385] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 10/19/2016] [Indexed: 12/20/2022] Open
Abstract
The arsenal for the treatment of metastatic melanoma is limited. A new approach to therapy using checkpoint blockade has improved overall survival in this patient population. Ipilimumab a CTLA-4 monoclonal antibody is a first in class drug that has pioneered this revolution. In this review, the authors provide an account of the different stages that led to the development of ipilimumab, its approval in the clinical setting for the treatment of advanced melanoma and ongoing investigations of combinatorial immune therapy.
Collapse
Affiliation(s)
- Paul Letendre
- Department of Hematology, Oncology & Blood & Marrow Transplantation & the Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, USA
| | - Varun Monga
- Department of Hematology, Oncology & Blood & Marrow Transplantation & the Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, USA
| | - Mohammed Milhem
- Department of Hematology, Oncology & Blood & Marrow Transplantation & the Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, USA
| | - Yousef Zakharia
- Department of Hematology, Oncology & Blood & Marrow Transplantation & the Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
203
|
Roh MR, Eliades P, Gupta S, Grant-Kels JM, Tsao H. Cutaneous melanoma in women. Int J Womens Dermatol 2017; 3:S11-S15. [PMID: 28492033 PMCID: PMC5419022 DOI: 10.1016/j.ijwd.2017.02.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2014] [Revised: 01/12/2015] [Accepted: 01/13/2015] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Gender disparity in melanoma outcome is consistently observed, suggesting that gender is as an important prognostic factor. However, the source of this gender disparity in melanoma remains unclear. OBJECTIVE This article reviews advances in our understanding of gender differences in melanoma and how such differences may contribute to outcomes. METHODS A broad literature search was conducted using the PubMed database, with search terms such as 'gender differences in melanoma' and 'sex differences in melanoma.' Additional articles were identified from cited references. RESULTS Herein, we address the gender-linked physiologic differences in skin and melanoma. We discuss the influence of estrogen on a woman's risk for melanoma and melanoma outcomes with regard to pregnancy, oral contraceptives, hormone replacement therapy, and UV tanning. CONCLUSIONS The published findings on gender disparities in melanoma have yielded many advances in our understanding of this disease. Biological, environmental, and behavioral factors may explain the observed gender difference in melanoma incidence and outcome. Further research will enable us to learn more about melanoma pathogenesis, with the goal of offering better treatments and preventative advice to our patients.
Collapse
Affiliation(s)
- Mi Ryung Roh
- Wellman Center for Photomedicine, Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, MA.,Department of Dermatology, Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Philip Eliades
- Wellman Center for Photomedicine, Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, MA.,Tufts University School of Medicine, Boston, MA
| | - Sameer Gupta
- Wellman Center for Photomedicine, Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Jane M Grant-Kels
- Dermatology Department, University of CT Health Center, Farmington, CT
| | - Hensin Tsao
- Wellman Center for Photomedicine, Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
204
|
Quinn C, Ma Q, Kudlac A, Palmer S, Barber B, Zhao Z. Relative Efficacy of Granulocyte-Macrophage Colony-Stimulating Factor, Dacarbazine, and Glycoprotein 100 in Metastatic Melanoma: An Indirect Treatment Comparison. Adv Ther 2017; 34:495-512. [PMID: 28000169 PMCID: PMC5331084 DOI: 10.1007/s12325-016-0464-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Advances in the treatment of metastatic melanoma have been achieved in recent years: immunotherapies and targeted therapies have demonstrated survival benefits over older agents such as granulocyte-macrophage colony-stimulating factor (GM-CSF), dacarbazine, and glycoprotein peptide vaccine (gp100) in pivotal phase 3 trials. It is important to compare therapies to guide the treatment decision-making process, and establishing the relationship between older agents can strengthen the networks of evidence for newer therapies. We report the outcome of an indirect comparison of GM-CSF, dacarbazine, and gp100 in metastatic melanoma through meta-analysis of absolute treatment effect. METHODS A systematic literature review identified trials for inclusion in the meta-analysis. A valid network meta-analysis was not feasible: treatment-specific meta-analysis was conducted. A published algorithm was used to adjust overall survival estimates from trials of GM-CSF, dacarbazine, and gp100 for heterogeneity in baseline prognostic factors. Survival estimates were compared in three patient groups: stage IIIB-IV M1c, stage IIIB-IV M1a, and stage IV M1b/c. RESULTS One trial of GM-CSF, four of dacarbazine, and one of gp100 were included in the analysis. After adjusting for differences in baseline prognostic factors, median overall survival (OS) in all patient groups was longer for those receiving GM-CSF than for those receiving dacarbazine or gp100. The observed survival over time for GM-CSF was similar to the adjusted survival for dacarbazine and greater than for gp100 in all patient groups. CONCLUSION The relative treatment effect of GM-CSF, dacarbazine, and gp100 has been reliably estimated by adjusting for differences in baseline prognostic factors. Results suggest that OS with GM-CSF is at least as good as with dacarbazine and greater than with gp100. Given the role of these agents as controls in phase 3 trials of new immunotherapies and targeted agents, these results can be used to contextualize the efficacy of newer therapies. FUNDING Amgen Inc.
Collapse
Affiliation(s)
| | | | | | - Stephen Palmer
- Centre for Health Economics, University of York, Heslington, UK
| | | | | |
Collapse
|
205
|
|
206
|
Willmes C, Kumar R, Becker JC, Fried I, Rachakonda PS, Poppe LM, Hesbacher S, Schadendorf D, Sucker A, Schrama D, Ugurel S. SERPINB1 expression is predictive for sensitivity and outcome of cisplatin-based chemotherapy in melanoma. Oncotarget 2017; 7:10117-32. [PMID: 26799424 PMCID: PMC4891108 DOI: 10.18632/oncotarget.6956] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 12/26/2015] [Indexed: 12/02/2022] Open
Abstract
Despite of highly effective new therapeutic strategies, chemotherapy still is an important treatment option in metastatic melanoma. Since predictors of chemotherapy response are rare, drugs and regimens are currently chosen arbitrarily. The present study was aimed at the identification of molecular markers predicting the outcome of chemotherapy in melanoma. Tumor biopsies from metastatic lesions were collected from 203 stage IV melanoma patients prior to chemotherapy onset and used for gene expression profiling (n = 6; marker identification set), quantitative real-time PCR (n = 127; validation set 1), and immunohistochemistry on tissue microarrays (n = 70; validation set 2). The results were correlated to the tumors' in-vitro chemosensitivity and to the patients' in-vivo chemotherapy outcome. SERPINB1 was found to correlate to the in-vitro sensitivity to cisplatin-containing chemotherapy regimens (p = 0.005). High SERPINB1 gene expression was associated with favorable tumor response (p = 0.012) and prolonged survival (p = 0.081) under cisplatin-based chemotherapy. High SERPINB1 protein expression in tumor tissue from cisplatin-treated patients was associated with a favorable survival (p = 0.011), and proved as an independent predictor of survival (p = 0.008) by multivariate analysis. We conclude, that SERPINB1 expression, although not functionally involved, is predictive for the outcome of cisplatin-based chemotherapy in melanoma, and thus may be useful to personalize melanoma chemotherapy.
Collapse
Affiliation(s)
- Christoph Willmes
- Department of Dermatology, University Hospital Würzburg, Würzburg, Germany
| | - Rajiv Kumar
- Division of Molecular Genetic Epidemiology, German Cancer Research Center, Heidelberg, Germany
| | - Jürgen C Becker
- Translational Skin Cancer Research, Deutsches Konsortium für Translationale Krebsforschung (DKTK), Essen, Germany.,Department of Dermatology, University Duisburg-Essen, Essen, Germany
| | - Isabella Fried
- Department of Dermatology, Medical University of Graz, Graz, Austria
| | | | - Lidia M Poppe
- Department of Dermatology, University Hospital Würzburg, Würzburg, Germany
| | - Sonja Hesbacher
- Department of Dermatology, University Hospital Würzburg, Würzburg, Germany
| | - Dirk Schadendorf
- Department of Dermatology, University Duisburg-Essen, Essen, Germany
| | - Antje Sucker
- Department of Dermatology, University Duisburg-Essen, Essen, Germany
| | - David Schrama
- Department of Dermatology, University Hospital Würzburg, Würzburg, Germany
| | - Selma Ugurel
- Department of Dermatology, University Hospital Würzburg, Würzburg, Germany.,Department of Dermatology, University Duisburg-Essen, Essen, Germany
| |
Collapse
|
207
|
Spath L, Ulivieri A, Lavra L, Fidanza L, Carlesimo M, Giubettini M, Narcisi A, Luciani E, Bucci B, Pisani D, Sciacchitano S, Bartolazzi A. Antiproliferative Effects of 1α-OH-vitD 3 in Malignant Melanoma: Potential Therapeutic implications. Sci Rep 2017; 7:40370. [PMID: 28074906 PMCID: PMC5225467 DOI: 10.1038/srep40370] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 12/06/2016] [Indexed: 02/06/2023] Open
Abstract
Early detection and surgery represent the mainstay of treatment for superficial melanoma, but for high risk lesions (Breslow’s thickness >0.75 mm) an effective adjuvant therapy is lacking. Vitamin D insufficiency plays a relevant role in cancer biology. The biological effects of 1α hydroxycholecalciferol on experimental melanoma models were investigated. 105 melanoma patients were checked for 25-hydroxycholecalciferol (circulating vitamin D) serum levels. Human derived melanoma cell lines and in vivo xenografts were used for studying 1α-hydroxycholecalciferol-mediated biological effects on cell proliferation and tumor growth. 99 out of 105 (94%) melanoma patients had insufficient 25-hydroxycholecalciferol serum levels. Interestingly among the six with vitamin D in the normal range, five had a diagnosis of in situ/microinvasive melanoma. Treatment with 1α-hydroxycholecalciferol induced antiproliferative effects on melanoma cells in vitro and in vivo, modulating the expression of cell cycle key regulatory molecules. Cell cycle arrest in G1 or G2 phase was invariably observed in vitamin D treated melanoma cells. The antiproliferative activity induced by 1α-hydroxycholecalciferol in experimental melanoma models, together with the discovery of insufficient 25-hydroxycholecalciferol serum levels in melanoma patients, provide the rationale for using vitamin D in melanoma adjuvant therapy, alone or in association with other therapeutic options.
Collapse
Affiliation(s)
- Lucia Spath
- Pathology Research Laboratory, Sant'Andrea University Hospital, via di Grottarossa 1035, 00189 Rome, Italy
| | - Alessandra Ulivieri
- Pathology Research Laboratory, Sant'Andrea University Hospital, via di Grottarossa 1035, 00189 Rome, Italy.,Laboratory of Biomedical Research, Niccolò Cusano University Foundation, via Don Carlo Gnocchi 3, 00166 Rome, Italy
| | - Luca Lavra
- Pathology Research Laboratory, Sant'Andrea University Hospital, via di Grottarossa 1035, 00189 Rome, Italy.,Laboratory of Biomedical Research, Niccolò Cusano University Foundation, via Don Carlo Gnocchi 3, 00166 Rome, Italy
| | - Laura Fidanza
- Dermatology Unit, Sant'Andrea University Hospital, via di Grottarossa 1035, 00189 Rome, Italy
| | - Marta Carlesimo
- Dermatology Unit, Sant'Andrea University Hospital, via di Grottarossa 1035, 00189 Rome, Italy
| | - Maria Giubettini
- Pathology Research Laboratory, Sant'Andrea University Hospital, via di Grottarossa 1035, 00189 Rome, Italy
| | - Alessandra Narcisi
- Dermatology Unit, Sant'Andrea University Hospital, via di Grottarossa 1035, 00189 Rome, Italy
| | - Emidio Luciani
- Pathology Research Laboratory, Sant'Andrea University Hospital, via di Grottarossa 1035, 00189 Rome, Italy
| | - Barbara Bucci
- Pathology Research Laboratory, Sant'Andrea University Hospital, via di Grottarossa 1035, 00189 Rome, Italy
| | - Daniela Pisani
- Internal Medicine Sant'Andrea University Hospital, via di Grottarossa 1035, 00189 Rome, Italy
| | - Salvatore Sciacchitano
- Laboratory of Biomedical Research, Niccolò Cusano University Foundation, via Don Carlo Gnocchi 3, 00166 Rome, Italy
| | - Armando Bartolazzi
- Pathology Research Laboratory, Sant'Andrea University Hospital, via di Grottarossa 1035, 00189 Rome, Italy.,Molecular and Cellular Tumor Pathology Laboratory, Cancer Center Karolinska, Karolinska Hospital, CCK R8:04, S-17176, Stockholm, Sweden
| |
Collapse
|
208
|
Melief SM, Visconti VV, Visser M, van Diepen M, Kapiteijn EHW, van den Berg JH, Haanen JBAG, Smit VTHBM, Oosting J, van der Burg SH, Verdegaal EME. Long-term Survival and Clinical Benefit from Adoptive T-cell Transfer in Stage IV Melanoma Patients Is Determined by a Four-Parameter Tumor Immune Signature. Cancer Immunol Res 2017; 5:170-179. [PMID: 28073773 DOI: 10.1158/2326-6066.cir-16-0288] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 12/09/2016] [Accepted: 12/11/2016] [Indexed: 11/16/2022]
Abstract
The presence of tumor-infiltrating immune cells is associated with longer survival and a better response to immunotherapy in early-stage melanoma, but a comprehensive study of the in situ immune microenvironment in stage IV melanoma has not been performed. We investigated the combined influence of a series of immune factors on survival and response to adoptive cell transfer (ACT) in stage IV melanoma patients. Metastases of 73 stage IV melanoma patients, 17 of which were treated with ACT, were studied with respect to the number and functional phenotype of lymphocytes and myeloid cells as well as for expression of galectins-1, -3, and -9. Single factors associated with better survival were identified using Kaplan-Meier curves and multivariate Cox regression analyses, and those factors were used for interaction analyses. The results were validated using The Cancer Genome Atlas database. We identified four parameters that were associated with a better survival: CD8+ T cells, galectin-9+ dendritic cells (DC)/DC-like macrophages, a high M1/M2 macrophage ratio, and the expression of galectin-3 by tumor cells. The presence of at least three of these parameters formed an independent positive prognostic factor for long-term survival. Patients displaying this four-parameter signature were found exclusively among patients responding to ACT and were the ones with sustained clinical benefit. Cancer Immunol Res; 5(2); 170-9. ©2017 AACR.
Collapse
Affiliation(s)
- Sara M Melief
- Department of Clinical Oncology, Leiden University Medical Center, Leiden, the Netherlands
| | - Valeria V Visconti
- Department of Clinical Oncology, Leiden University Medical Center, Leiden, the Netherlands
| | - Marten Visser
- Department of Clinical Oncology, Leiden University Medical Center, Leiden, the Netherlands
| | - Merel van Diepen
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Ellen H W Kapiteijn
- Department of Clinical Oncology, Leiden University Medical Center, Leiden, the Netherlands
| | - Joost H van den Berg
- Division of Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - John B A G Haanen
- Division of Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Vincent T H B M Smit
- Department of Pathology, Leiden University Medical Center, Leiden, the Netherlands
| | - Jan Oosting
- Bioinformatics Center of Expertise, Leiden University Medical Center, Leiden, the Netherlands
| | - Sjoerd H van der Burg
- Department of Clinical Oncology, Leiden University Medical Center, Leiden, the Netherlands
| | - Els M E Verdegaal
- Department of Clinical Oncology, Leiden University Medical Center, Leiden, the Netherlands.
| |
Collapse
|
209
|
Wang X, Feng Y, Bajaj G, Gupta M, Agrawal S, Yang A, Park J, Lestini B, Roy A. Quantitative Characterization of the Exposure-Response Relationship for Cancer Immunotherapy: A Case Study of Nivolumab in Patients With Advanced Melanoma. CPT Pharmacometrics Syst Pharmacol 2017; 6:40-48. [PMID: 28019090 PMCID: PMC5270290 DOI: 10.1002/psp4.12133] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 09/02/2016] [Indexed: 12/14/2022] Open
Abstract
To inform the benefit-risk assessment of nivolumab in patients with advanced melanoma, analyses of efficacy and safety exposure-response (E-R) relationships were conducted with data from patients with advanced melanoma enrolled in two clinical studies (phase I and phase III) who received nivolumab 0.1-10.0 mg/kg every 2 weeks. E-R efficacy analyses were performed by relating the nivolumab time-averaged concentration after the first dose (Cavg1 ) to two endpoints: RECIST objective response (OR) and overall survival (OS). E-R safety analyses characterized the relationship between nivolumab Cavg1 and the hazard of all-causality adverse events leading to discontinuation or death (AE-DC/D). Nivolumab exposure represented by Cavg1 was not a significant predictor of OR, OS, or the hazard of AE-DC/D. E-R efficacy and safety relationships were relatively flat over the exposure range.
Collapse
Affiliation(s)
- X Wang
- Clinical Pharmacology and PharmacometricsBristol‐Myers SquibbPrincetonNew JerseyUSA
| | - Y Feng
- Clinical Pharmacology and PharmacometricsBristol‐Myers SquibbPrincetonNew JerseyUSA
| | - G Bajaj
- Clinical Pharmacology and PharmacometricsBristol‐Myers SquibbPrincetonNew JerseyUSA
| | - M Gupta
- Clinical Pharmacology and PharmacometricsBristol‐Myers SquibbPrincetonNew JerseyUSA
| | - S Agrawal
- Clinical Pharmacology and PharmacometricsBristol‐Myers SquibbPrincetonNew JerseyUSA
| | - A Yang
- Oncology Global Clinical Research, Bristol‐Myers SquibbPrincetonNew JerseyUSA
| | - J‐S Park
- Global Biometric Sciences, Bristol‐Myers SquibbPrincetonNew JerseyUSA
| | - B Lestini
- Oncology Global Clinical Research, Bristol‐Myers SquibbPrincetonNew JerseyUSA
| | - A Roy
- Clinical Pharmacology and PharmacometricsBristol‐Myers SquibbPrincetonNew JerseyUSA
| |
Collapse
|
210
|
Jochems A, Schouwenburg MG, Leeneman B, Franken MG, van den Eertwegh AJM, Haanen JBAG, Gelderblom H, Uyl-de Groot CA, Aarts MJB, van den Berkmortel FWPJ, Blokx WAM, Cardous-Ubbink MC, Groenewegen G, de Groot JWB, Hospers GAP, Kapiteijn E, Koornstra RH, Kruit WH, Louwman MW, Piersma D, van Rijn RS, Ten Tije AJ, Vreugdenhil G, Wouters MWJM, van der Hoeven JJM. Dutch Melanoma Treatment Registry: Quality assurance in the care of patients with metastatic melanoma in the Netherlands. Eur J Cancer 2016; 72:156-165. [PMID: 28030784 DOI: 10.1016/j.ejca.2016.11.021] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 10/29/2016] [Accepted: 11/15/2016] [Indexed: 11/29/2022]
Abstract
BACKGROUND In recent years, the treatment of metastatic melanoma has changed dramatically due to the development of immune checkpoint and mitogen-activated protein (MAP) kinase inhibitors. A population-based registry, the Dutch Melanoma Treatment Registry (DMTR), was set up in July 2013 to assure the safety and quality of melanoma care in the Netherlands. This article describes the design and objectives of the DMTR and presents some results of the first 2 years of registration. METHODS The DMTR documents detailed information on all Dutch patients with unresectable stage IIIc or IV melanoma. This includes tumour and patient characteristics, treatment patterns, clinical outcomes, quality of life, healthcare utilisation, informal care and productivity losses. These data are used for clinical auditing, increasing the transparency of melanoma care, providing insights into real-world cost-effectiveness and creating a platform for research. RESULTS Within 1 year, all melanoma centres were participating in the DMTR. The quality performance indicators demonstrated that the BRAF inhibitors and ipilimumab have been safely introduced in the Netherlands with toxicity rates that were consistent with the phase III trials conducted. The median overall survival of patients treated with systemic therapy was 10.1 months (95% confidence interval [CI] 9.1-11.1) in the first registration year and 12.7 months (95% CI 11.6-13.7) in the second year. CONCLUSION The DMTR is the first comprehensive multipurpose nationwide registry and its collaboration with all stakeholders involved in melanoma care reflects an integrative view of cancer management. In future, the DMTR will provide insights into challenging questions regarding the definition of possible subsets of patients who benefit most from the new drugs.
Collapse
Affiliation(s)
- Anouk Jochems
- Department of Medical Oncology, Leiden University Medical Centre, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Maartje G Schouwenburg
- Department of Medical Oncology, Leiden University Medical Centre, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Brenda Leeneman
- Institute for Medical Technology Assessment, Erasmus University, Burgemeester Oudlaan 50, 3062 PA, Rotterdam, The Netherlands
| | - Margreet G Franken
- Institute for Medical Technology Assessment, Erasmus University, Burgemeester Oudlaan 50, 3062 PA, Rotterdam, The Netherlands
| | - Alfons J M van den Eertwegh
- Department of Medical Oncology, VU University Medical Centre, De Boelelaan 1118, 1081 HZ, Amsterdam, The Netherlands
| | - John B A G Haanen
- Department of Medical Oncology, Netherlands Cancer Institute - Antoni van Leeuwenhoek hospital, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Hans Gelderblom
- Department of Medical Oncology, Leiden University Medical Centre, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Carin A Uyl-de Groot
- Institute for Medical Technology Assessment, Erasmus University, Burgemeester Oudlaan 50, 3062 PA, Rotterdam, The Netherlands
| | - Maureen J B Aarts
- Department of Medical Oncology, Maastricht University Medical Centre, P. Debyelaan 25, 6229 HX, Maastricht, The Netherlands
| | | | - Willeke A M Blokx
- Department of Pathology, Radboud University Medical Centre, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, The Netherlands
| | - Mathilde C Cardous-Ubbink
- Netherlands Comprehensive Cancer Organisation, Godebaldkwartier 419, 3511 DT, Utrecht, The Netherlands
| | - Gerard Groenewegen
- Department of Medical Oncology, University Medical Centre Utrecht, Heidelberglaan 100, 3582, CX, Utrecht, The Netherlands
| | - Jan Willem B de Groot
- Department of Medical Oncology, Isala, Dokter van Heesweg 2, 8025 AB, Zwolle, The Netherlands
| | - Geke A P Hospers
- Department of Medical Oncology, University Medical Centre Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Ellen Kapiteijn
- Department of Medical Oncology, Leiden University Medical Centre, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Rutger H Koornstra
- Department of Medical Oncology, Radboud University Medical Centre, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, The Netherlands
| | - Wim H Kruit
- Department of Medical Oncology, Erasmus MC Cancer Institute, 's-Gravendijkwal 230, 3015 CE, Rotterdam, The Netherlands
| | - Marieke W Louwman
- Netherlands Comprehensive Cancer Organisation, Godebaldkwartier 419, 3511 DT, Utrecht, The Netherlands
| | - Djura Piersma
- Department of Internal Medicine, Medisch Spectrum Twente, Koningsplein 1, 7512 KZ, Enschede, The Netherlands
| | - Rozemarijn S van Rijn
- Department of Internal Medicine, Medical Centre Leeuwarden, Henri Dunantweg 2, 8934 AD, Leeuwarden, The Netherlands
| | - Albert J Ten Tije
- Department of Internal Medicine, Amphia Hospital, Molengracht 21, 4818 CK, Breda, The Netherlands
| | - Gerard Vreugdenhil
- Department of Internal Medicine, Maxima Medical Centre, De Run 4600, 5504 DB, Eindhoven, The Netherlands
| | - Michel W J M Wouters
- Department of Surgical Oncology, Netherlands Cancer Institute - Antoni van Leeuwenhoek hospital, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Jacobus J M van der Hoeven
- Department of Medical Oncology, Leiden University Medical Centre, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands.
| |
Collapse
|
211
|
Ernst DS, Petrella T, Joshua AM, Hamou A, Thabane M, Vantyghem S, Gwadry-Sridhar F. Burden of illness for metastatic melanoma in Canada, 2011-2013. ACTA ACUST UNITED AC 2016; 23:e563-e570. [PMID: 28050145 DOI: 10.3747/co.23.3161] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
BACKGROUND Detailed epidemiology for patients with advanced metastatic melanoma in Canada is not well characterized. We conducted an analysis of patients with this disease in the province of Ontario, with the aim being to study the presentation, disease characteristics and course, and treatment patterns for malignant melanoma. METHODS In this Canadian observational prospective and retrospective study of patients with malignant melanoma, we used data collected in the Canadian Melanoma Research Network (cmrn) Patient Registry. We identified patients who were seen at 1 of 3 cancer treatment centres between April 2011 and 30 April 2013. Patient data from 2011 and 2012 were collected retrospectively using chart records and existing registry data. Starting January 2013, data were collected prospectively. Variables investigated included age, sex, initial stage, histology, mutation type, time to recurrence, sites of metastases, resectability, and previous therapies. RESULTS A cohort of 810 patients with melanoma was identified from the cmrn registry. Mean age was 58.7 years, and most patients were men (60% vs. 40%). Factors affecting survival included unresectable or metastatic melanoma, initial stage at diagnosis, presence of brain metastasis, and BRAF mutation status. The proportion of surviving patients decreased with higher initial disease stages. CONCLUSIONS Using registry data, we were able to determine the detailed epidemiology of patients with melanoma in the Canadian province of Ontario, validating the comprehensive and detailed information that can be obtained from registry data.
Collapse
Affiliation(s)
- D S Ernst
- London Regional Cancer Program, London Health Sciences Centre, London, ON
| | - T Petrella
- Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, ON
| | - A M Joshua
- Princess Margaret Cancer Centre, Toronto, ON
| | - A Hamou
- Western University, London, ON
| | - M Thabane
- Novartis Pharmaceuticals Canada Inc., Dorval, QC
| | - S Vantyghem
- Novartis Pharmaceuticals Canada Inc., Dorval, QC
| | | |
Collapse
|
212
|
Gee MS, Ghazani AA, Haq R, Wargo JA, Sebas M, Sullivan RJ, Lee H, Weissleder R. Point of care assessment of melanoma tumor signaling and metastatic burden from μNMR analysis of tumor fine needle aspirates and peripheral blood. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2016; 13:821-828. [PMID: 27993725 DOI: 10.1016/j.nano.2016.12.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2016] [Revised: 11/17/2016] [Accepted: 12/05/2016] [Indexed: 11/29/2022]
Abstract
This study evaluates μNMR technology for molecular profiling of tumor fine needle aspirates and peripheral blood of melanoma patients. In vitro assessment of melanocyte (MART-1, HMB45) and MAP kinase signaling (pERK, pS6K) molecule expression was performed in human cell lines, while clinical validation was performed in an IRB-approved study of melanoma patients undergoing biopsy and blood sampling. Tumor FNA and blood specimens were compared with BRAF genetic analysis and cross-sectional imaging. μNMR in vitro analysis showed increased expression of melanocyte markers in melanoma cells as well as increased expression of phosphorylated MAP kinase targets in BRAF-mutant melanoma cells. Melanoma patient FNA samples showed increased pERK and pS6K levels in BRAF mutant compared with BRAF WT melanomas, with μNMR blood circulating tumor cell level increased with higher metastatic burden visible on imaging. These results indicate that μNMR technology provides minimally invasive point-of-care evaluation of tumor signaling and metastatic burden in melanoma patients.
Collapse
Affiliation(s)
- Michael S Gee
- Department of Radiology, Massachusetts General Hospital, Boston, MA 02114.,Center for Systems Biology, Massachusetts General Hospital, Boston, MA 02114
| | - Arezou A Ghazani
- Center for Systems Biology, Massachusetts General Hospital, Boston, MA 02114
| | - Rizwan Haq
- Department of Radiology, Massachusetts General Hospital, Boston, MA 02114
| | - Jennifer A Wargo
- Department of Radiology, Massachusetts General Hospital, Boston, MA 02114
| | - Matthew Sebas
- Center for Systems Biology, Massachusetts General Hospital, Boston, MA 02114
| | - Ryan J Sullivan
- Center for Melanoma, Massachusetts General Hospital Cancer Center, Boston, MA 02114
| | - Hakho Lee
- Center for Systems Biology, Massachusetts General Hospital, Boston, MA 02114
| | - Ralph Weissleder
- Department of Radiology, Massachusetts General Hospital, Boston, MA 02114.,Center for Systems Biology, Massachusetts General Hospital, Boston, MA 02114
| |
Collapse
|
213
|
Bohensky MA, Pasupathi K, Gorelik A, Kim H, Harrison JP, Liew D. A Cost-Effectiveness Analysis of Nivolumab Compared with Ipilimumab for the Treatment of BRAF Wild-Type Advanced Melanoma in Australia. VALUE IN HEALTH : THE JOURNAL OF THE INTERNATIONAL SOCIETY FOR PHARMACOECONOMICS AND OUTCOMES RESEARCH 2016; 19:1009-1015. [PMID: 27987627 DOI: 10.1016/j.jval.2016.05.013] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Revised: 05/09/2016] [Accepted: 05/18/2016] [Indexed: 05/25/2023]
Abstract
PURPOSE The aim of this study was to evaluate the cost-effectiveness of nivolumab versus ipilimumab for the treatment of previously untreated patients with BRAF-advanced melanoma (BRAF-AM) from an Australian health system perspective. METHODS A state-transition Markov model was constructed to simulate the progress of Australian patients with BRAF-AM. The model had a 10-year time horizon with outcomes discounted at 5% annually. For the nivolumab group, risks of progression and death were based on those observed in the nivolumab arm of a phase III trial (nivolumab vs. dacarbazine). Progression-free survival and overall survival were extrapolated using parametric survival modeling with a log-logistic distribution. In the absence of head-to-head evidence, overall survival and progression-free survival for ipilimumab were estimated on the basis of an indirect comparison using published data. Costs of managing AM were estimated from a survey of Australian clinicians. The cost of ipilimumab was based on the reimbursement price in Australia. The cost of nivolumab was based on expected reimbursement prices in Australia. Quality-of-life data were obtained within the trial using the EuroQol five-dimensional questionnaire. RESULTS Compared with ipilimumab, nivolumab therapy over 10 years was estimated to yield 1.58 life-years and 1.30 quality-adjusted life-years per person, at a (discounted) net cost of US $39,039 per person. The incremental cost-effectiveness ratios for nivolumab compared with ipilimumab were US $25,101 per year of life saved and $30,475 per quality-adjusted life-year saved. CONCLUSIONS Nivolumab is a cost-effective means of preventing downstream mortality and morbidity in patients with AM compared with ipilimumab in the Australian setting.
Collapse
Affiliation(s)
- Megan A Bohensky
- Melbourne EpiCentre, Royal Melbourne Hospital, and the University of Melbourne, Parkville, Victoria, Australia.
| | - Kumar Pasupathi
- Melbourne EpiCentre, Royal Melbourne Hospital, and the University of Melbourne, Parkville, Victoria, Australia
| | - Alexandra Gorelik
- Melbourne EpiCentre, Royal Melbourne Hospital, and the University of Melbourne, Parkville, Victoria, Australia
| | - Hansoo Kim
- Bristol-Myers Squibb Australia, Mulgrave, Victoria, Australia
| | | | - Danny Liew
- Melbourne EpiCentre, Royal Melbourne Hospital, and the University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
214
|
Eichler H, Bloechl‐Daum B, Bauer P, Bretz F, Brown J, Hampson LV, Honig P, Krams M, Leufkens H, Lim R, Lumpkin MM, Murphy MJ, Pignatti F, Posch M, Schneeweiss S, Trusheim M, Koenig F. "Threshold-crossing": A Useful Way to Establish the Counterfactual in Clinical Trials? Clin Pharmacol Ther 2016; 100:699-712. [PMID: 27650716 PMCID: PMC5114686 DOI: 10.1002/cpt.515] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Revised: 09/15/2016] [Accepted: 09/16/2016] [Indexed: 12/15/2022]
Abstract
A central question in the assessment of benefit/harm of new treatments is: how does the average outcome on the new treatment (the factual) compare to the average outcome had patients received no treatment or a different treatment known to be effective (the counterfactual)? Randomized controlled trials (RCTs) are the standard for comparing the factual with the counterfactual. Recent developments necessitate and enable a new way of determining the counterfactual for some new medicines. For select situations, we propose a new framework for evidence generation, which we call "threshold-crossing." This framework leverages the wealth of information that is becoming available from completed RCTs and from real world data sources. Relying on formalized procedures, information gleaned from these data is used to estimate the counterfactual, enabling efficacy assessment of new drugs. We propose future (research) activities to enable "threshold-crossing" for carefully selected products and indications in which RCTs are not feasible.
Collapse
Affiliation(s)
- H‐G Eichler
- European Medicines AgencyLondonUnited Kingdom
| | - B Bloechl‐Daum
- Department of Clinical PharmacologyMedical University of ViennaViennaAustria
| | - P Bauer
- Section for Medical Statistics, Center for Medical Statistics, Informatics, and Intelligent SystemsMedical University of ViennaViennaAustria
| | | | - J Brown
- Harvard Medical School/Harvard Pilgrim Health Care InstituteHartfordConnecticutUSA
| | - LV Hampson
- Lancaster UniversityLancasterUnited Kingdom
| | | | - M Krams
- Janssen Pharmaceutical CompaniesRaritanNew JerseyUSA
| | - H Leufkens
- Medicines Evaluation Board, UtrechtUniversity of UtrechtUtrechtThe Netherlands
| | - R Lim
- Health CanadaOttawaOntarioCanada
| | - MM Lumpkin
- Bill and Melinda Gates FoundationSeattleWashingtonUSA
| | - MJ Murphy
- Project Data SphereDurhamNorth CarolinaUSA
| | - F Pignatti
- European Medicines AgencyLondonUnited Kingdom
| | - M Posch
- Section for Medical Statistics, Center for Medical Statistics, Informatics, and Intelligent SystemsMedical University of ViennaViennaAustria
| | - S Schneeweiss
- Brigham and Women's Hospital and Harvard Medical SchoolBostonMassachusettsUSA
| | - M Trusheim
- MIT Sloan School of ManagementCambridgeMassachusettsUSA
| | - F Koenig
- Section for Medical Statistics, Center for Medical Statistics, Informatics, and Intelligent SystemsMedical University of ViennaViennaAustria
| |
Collapse
|
215
|
Rapoport BL, Vorobiof DA, Dreosti LM, Nosworthy A, McAdam G, Jordaan JP, Miller-Jansön H, de Necker M, de Beer JC, Duvenhage H. Ipilimumab in Pretreated Patients With Advanced Malignant Melanoma: Results of the South African Expanded-Access Program. J Glob Oncol 2016; 3:515-523. [PMID: 29094091 PMCID: PMC5646890 DOI: 10.1200/jgo.2016.006544] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Purpose The primary objective of this study was to evaluate 1- and 2-year survival rates and durable remissions in pretreated patients with advanced (unresectable or metastatic) malignant melanoma treated with ipilimumab in a South African expanded-access program (SA-EAP). Patients and Methods This multicenter, retrospective study obtained data from pretreated patients with advanced malignant melanoma who were eligible for the ipilimumab SA-EAP. Ipilimumab was administered at a dose of 3 mg/kg intravenously every 3 weeks for four cycles to adults with advanced melanoma for whom at least one line of treatment for metastatic disease had failed. Data from the medical records of 108 patients treated within the SA-EAP were collected and statistically analyzed to determine overall (OS) and progression-free survival (PFS) at 1 and 2 years. Results In the population of 108 patients, a median OS of 8.98 months (95% CI, 7.47 to 10.79 months) was observed. One-year OS was 36% (95% CI, 26% to 45%), and 2-year survival was observed as 20% (95% CI, 12% to 27%). The median survival without progression (ie, PFS) was 3.44 months (95% CI, 2.98 to 4.16 months), and 1- and 2-year PFS were 22% (95% CI, 14% to 29%) and 14% (95% CI, 8% to 21%), respectively. The longest recorded survival was 3.4 years. No independent prognostic variables were identified to predict for OS by multivariate Cox proportional hazards model. Conclusion In this multicenter South African setting, ipilimumab at a dose of 3 mg/kg was an effective treatment with long-term OS in a subset of patients with pretreated advanced malignant melanoma.
Collapse
Affiliation(s)
- Bernardo L Rapoport
- , Medical Oncology Centre of Rosebank; , Sandton Oncology Center; , Wits Oncology Donald Gordon Medical Center; , Bristol-Myers Squibb South Africa, Johannesburg; , University of Pretoria, Pretoria; , Rondebosch Oncology Medical Center, Cape Town; , Westridge Oncology Center, Durban; and , , and , HEXOR, Midrand, South Africa
| | - Daniel A Vorobiof
- , Medical Oncology Centre of Rosebank; , Sandton Oncology Center; , Wits Oncology Donald Gordon Medical Center; , Bristol-Myers Squibb South Africa, Johannesburg; , University of Pretoria, Pretoria; , Rondebosch Oncology Medical Center, Cape Town; , Westridge Oncology Center, Durban; and , , and , HEXOR, Midrand, South Africa
| | - Lydia M Dreosti
- , Medical Oncology Centre of Rosebank; , Sandton Oncology Center; , Wits Oncology Donald Gordon Medical Center; , Bristol-Myers Squibb South Africa, Johannesburg; , University of Pretoria, Pretoria; , Rondebosch Oncology Medical Center, Cape Town; , Westridge Oncology Center, Durban; and , , and , HEXOR, Midrand, South Africa
| | - Adam Nosworthy
- , Medical Oncology Centre of Rosebank; , Sandton Oncology Center; , Wits Oncology Donald Gordon Medical Center; , Bristol-Myers Squibb South Africa, Johannesburg; , University of Pretoria, Pretoria; , Rondebosch Oncology Medical Center, Cape Town; , Westridge Oncology Center, Durban; and , , and , HEXOR, Midrand, South Africa
| | - Georgina McAdam
- , Medical Oncology Centre of Rosebank; , Sandton Oncology Center; , Wits Oncology Donald Gordon Medical Center; , Bristol-Myers Squibb South Africa, Johannesburg; , University of Pretoria, Pretoria; , Rondebosch Oncology Medical Center, Cape Town; , Westridge Oncology Center, Durban; and , , and , HEXOR, Midrand, South Africa
| | - Johan P Jordaan
- , Medical Oncology Centre of Rosebank; , Sandton Oncology Center; , Wits Oncology Donald Gordon Medical Center; , Bristol-Myers Squibb South Africa, Johannesburg; , University of Pretoria, Pretoria; , Rondebosch Oncology Medical Center, Cape Town; , Westridge Oncology Center, Durban; and , , and , HEXOR, Midrand, South Africa
| | - Helen Miller-Jansön
- , Medical Oncology Centre of Rosebank; , Sandton Oncology Center; , Wits Oncology Donald Gordon Medical Center; , Bristol-Myers Squibb South Africa, Johannesburg; , University of Pretoria, Pretoria; , Rondebosch Oncology Medical Center, Cape Town; , Westridge Oncology Center, Durban; and , , and , HEXOR, Midrand, South Africa
| | - Margreet de Necker
- , Medical Oncology Centre of Rosebank; , Sandton Oncology Center; , Wits Oncology Donald Gordon Medical Center; , Bristol-Myers Squibb South Africa, Johannesburg; , University of Pretoria, Pretoria; , Rondebosch Oncology Medical Center, Cape Town; , Westridge Oncology Center, Durban; and , , and , HEXOR, Midrand, South Africa
| | - Janetta C de Beer
- , Medical Oncology Centre of Rosebank; , Sandton Oncology Center; , Wits Oncology Donald Gordon Medical Center; , Bristol-Myers Squibb South Africa, Johannesburg; , University of Pretoria, Pretoria; , Rondebosch Oncology Medical Center, Cape Town; , Westridge Oncology Center, Durban; and , , and , HEXOR, Midrand, South Africa
| | - Hennie Duvenhage
- , Medical Oncology Centre of Rosebank; , Sandton Oncology Center; , Wits Oncology Donald Gordon Medical Center; , Bristol-Myers Squibb South Africa, Johannesburg; , University of Pretoria, Pretoria; , Rondebosch Oncology Medical Center, Cape Town; , Westridge Oncology Center, Durban; and , , and , HEXOR, Midrand, South Africa
| |
Collapse
|
216
|
Ramelyte E, Schindler SA, Dummer R. The safety of anti PD-1 therapeutics for the treatment of melanoma. Expert Opin Drug Saf 2016; 16:41-53. [PMID: 27737598 DOI: 10.1080/14740338.2016.1248402] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
INTRODUCTION The introduction of immunotherapies into clinical practice has substantially improved the prognosis of metastatic melanoma patients as well as patients suffering from other cancers. The two FDA-approved checkpoint inhibitors against PD-1 (nivolumab and pembrolizumab) have been shown to significantly improve patient survival while being less toxic than previous treatment options. Areas covered: The current scientific literature on safety and adverse events (AEs) related to anti-PD-1 therapies has been investigated with special attention to case reports and to the latest results announced at the major clinical cancer and melanoma meetings, including ASCO (American Society of Clinical Oncology), ESMO (European Society of medical Oncology) and EADO (European Association of Dermato-Oncology) annual meetings. Expert opinion: Even though anti-PD-1 therapies are better tolerated than conventional chemo- or other immune-therapies, they still induce a plethora of AEs. Given the mechanism of action, it is supposed that most if not all of them are immune related. Fortunately, the majority are mild and manageable. However, due to the increase in patients' life expectancy, there is a substantial need to understand and prevent severe cutaneous, pulmonary, neurological and other AEs which have major impact on the quality of life. The safety profile after long term use of these medications is still unclear. In addition, non-steroid based immune interventions to control autoimmunity are still to be developed.
Collapse
Affiliation(s)
- Egle Ramelyte
- a Department of Dermatology , University Hospital Zurich , Zurich , Switzerland.,b Centre of Dermatovenereology , Vilnius University Hospital Santariskiu klinikos , Vilnius , Lithuania
| | - Sabrina A Schindler
- a Department of Dermatology , University Hospital Zurich , Zurich , Switzerland
| | - Reinhard Dummer
- a Department of Dermatology , University Hospital Zurich , Zurich , Switzerland
| |
Collapse
|
217
|
Kyte JA, Aamdal S, Dueland S, Sæbøe-Larsen S, Inderberg EM, Madsbu UE, Skovlund E, Gaudernack G, Kvalheim G. Immune response and long-term clinical outcome in advanced melanoma patients vaccinated with tumor-mRNA-transfected dendritic cells. Oncoimmunology 2016; 5:e1232237. [PMID: 27999747 DOI: 10.1080/2162402x.2016.1232237] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 08/30/2016] [Accepted: 08/31/2016] [Indexed: 01/18/2023] Open
Abstract
The most effective anticancer immune responses are probably directed against patient-specific neoantigens. We have developed a melanoma vaccine targeting this individual mutanome based on dendritic cells (DCs) loaded with autologous tumor-mRNA. Here, we report a phase I/II trial evaluating toxicity, immune response and clinical outcome in 31 metastatic melanoma patients. The first cohort (n = 22) received the vaccine without any adjuvant; the next cohort (n = 9) received adjuvant IL2. Each subject received four weekly intranodal or intradermal injections, followed by optional monthly vaccines. Immune response was evaluated by delayed-type hypersensitivity (DTH), T cell proliferation and cytokine assays. Data were collected for 10 y after inclusion of the last patient. No serious adverse events were detected. In the intention-to-treat-cohort, we demonstrated significantly superior survival compared to matched controls from a benchmark meta-analysis (1 y survival 43% vs. 24%, 2 y 23% vs. 6.6%). A tumor-specific immune response was demonstrated in 16/31 patients. The response rate was higher after intradermal than intranodal vaccination (80% vs. 38%). Immune responders had improved survival compared to non-responders (median 14 mo vs. 6 mo; p = 0.030), and all eight patients surviving >20 mo were immune responders. In addition to the tumor-specific response, most patients developed a response against autologous DC antigens. The cytokine profile was polyfunctional and did not follow a Th1/Th2 dichotomy. We conclude that the favorable safety profile and evidence of a possible survival benefit warrant further studies of the RNA/DC vaccine. The vaccine appears insufficient as monotherapy, but there is a strong rationale for combination with checkpoint modulators.
Collapse
Affiliation(s)
- Jon Amund Kyte
- Department for Cell Therapy, Radiumhospitalet, Oslo University Hospital, Oslo, Norway; The Clinical Trial Unit, Radiumhospitalet, Oslo University Hospital, Oslo, Norway; Department of Immunology, Radiumhospitalet, Oslo University Hospital, Oslo, Norway
| | - Steinar Aamdal
- The Clinical Trial Unit, Radiumhospitalet, Oslo University Hospital , Oslo, Norway
| | - Svein Dueland
- The Clinical Trial Unit, Radiumhospitalet, Oslo University Hospital , Oslo, Norway
| | - Stein Sæbøe-Larsen
- Department for Cell Therapy, Radiumhospitalet, Oslo University Hospital , Oslo, Norway
| | - Else Marit Inderberg
- Department for Cell Therapy, Radiumhospitalet, Oslo University Hospital , Oslo, Norway
| | - Ulf Erik Madsbu
- Department for Radiology, Radiumhospitalet, Oslo University Hospital , Oslo, Norway
| | - Eva Skovlund
- Department of Public Health and General Practice, NTNU , Trondheim, Norway
| | - Gustav Gaudernack
- Department of Immunology, Radiumhospitalet, Oslo University Hospital , Oslo, Norway
| | - Gunnar Kvalheim
- Department for Cell Therapy, Radiumhospitalet, Oslo University Hospital , Oslo, Norway
| |
Collapse
|
218
|
Yan H, Hou X, Li T, Zhao L, Yuan X, Fu H, Zhu R. CD4+ T cell-mediated cytotoxicity eliminates primary tumor cells in metastatic melanoma through high MHC class II expression and can be enhanced by inhibitory receptor blockade. Tumour Biol 2016; 37:10.1007/s13277-016-5456-5. [PMID: 27709550 DOI: 10.1007/s13277-016-5456-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 09/23/2016] [Indexed: 01/02/2023] Open
Abstract
Metastatic melanoma is a rapidly progressing disease with high mortality rate and limited treatment options. Immunotherapy based on tumor-targeting cytotoxic T cell responses represents a promising strategy. To assist in its development, we examined the possibility and efficacy of using CD4+ cytotoxic T cells. The regulatory mechanisms controlling CD4+ T cell-mediated cytotoxicity were also investigated. We found that naturally occurring granzyme B and perforin-expressing CD4+ cytotoxic T cells can be recovered from metastatic melanoma patients at significantly elevated frequencies compared to those from healthy controls. These CD4+ cytotoxic T cells were also capable of killing autologous tumor cells harvested from metastatic melanoma, independent of CD8+ T cells or any other cell types. However, several restricting factors were observed. First, the cytolytic activity by CD4+ T cells required high MHC class II expression on melanoma cells, which was not satisfied in a subset of melanomas. Second, the granzyme B and perforin release by activated CD4+ cytotoxic T cells was reduced after coculturing with autologous melanoma cells, characterized by low LAMP-1 expression and low granzyme B and perforin secretion in the supernatant. This suggested that inhibitory mechanisms were present to suppress CD4+ cytotoxic T cells. Indeed, blockade of PD-1 and CTLA-4 had increased the cytolytic activity of CD4+ T cells but was only effective in MHC class II high but not MHC class II low melanomas. Together, our study showed that CD4+ T cell-mediated cytotoxicity could eliminate primary melanoma cells but the efficacy depended on MHC class II expression.
Collapse
Affiliation(s)
- Hongxia Yan
- Department of Dermatology, The First People's Hospital of Jining City, 6 Jiankang Road, Jining, Shangdong, 272011, China
| | - Xianglian Hou
- Department of Supply and Services, Jiaxiang County People's Hospital, Jiaxiang County, Jining, Shangdong, 272400, China
| | - Tianhang Li
- Department of Dermatology, The First People's Hospital of Jining City, 6 Jiankang Road, Jining, Shangdong, 272011, China.
| | - Li Zhao
- Department of Dermatology, The First People's Hospital of Jining City, 6 Jiankang Road, Jining, Shangdong, 272011, China
| | - Xiaozhou Yuan
- DICAT Biomedical Computation Centre, Vancouver, BC, Canada
| | - Hongjun Fu
- Department of Dermatology, The First People's Hospital of Jining City, 6 Jiankang Road, Jining, Shangdong, 272011, China
| | - Ruijie Zhu
- Department of Dermatology, The First People's Hospital of Jining City, 6 Jiankang Road, Jining, Shangdong, 272011, China
| |
Collapse
|
219
|
Wieder T, Brenner E, Braumüller H, Röcken M. Immunotherapy of melanoma: efficacy and mode of action. J Dtsch Dermatol Ges 2016; 14:28-37. [PMID: 26713633 DOI: 10.1111/ddg.12819] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Forty years of research have brought about the development of antibodies that induce effective antitumor immune responses through sustained activation of the immune system. These "immune checkpoint inhibitors" are directed against immune inhibitory molecules, such as cytotoxic T lymphocyte antigen 4 (CTLA-4), programmed death 1 (PD-1) or programmed death ligand 1 (PD-L1). Disruption of the PD-1/PD-L1 interaction improves the intermediate-term prognosis even in patients with advanced stage IV melanoma. One and a half years after treatment initiation, 30-60 % of these patients are still alive. While cancer immunotherapies usually do not eradicate metastases completely, they do cause a regression by 20-80 %. It is well established that the immune system is able to kill tumor cells, and this has also been demonstrated for immunotherapies. Preclinical data, however, has shown that anti-cancer immunity is not limited to killing cancer cells. Thus, through interferon gamma and tumor necrosis factor, the immune system is able to induce stable tumor growth arrest, referred to as senescence. Ensuring patient survival by long-term stabilization of metastatic growth will therefore become a central goal of antitumor immunotherapies. This therapeutic approach is effective in melanoma and non-small-cell lung cancer. Once immunotherapies also have an indication for common cancer types, drug prices will have to drop considerably in order to be able to keep them available to those dependent on such therapies.
Collapse
Affiliation(s)
- Thomas Wieder
- Department of Dermatology, Eberhard Karls University, Tübingen, Germany
| | - Ellen Brenner
- Department of Dermatology, Eberhard Karls University, Tübingen, Germany
| | - Heidi Braumüller
- Department of Dermatology, Eberhard Karls University, Tübingen, Germany
| | - Martin Röcken
- Department of Dermatology, Eberhard Karls University, Tübingen, Germany
| |
Collapse
|
220
|
JUN dependency in distinct early and late BRAF inhibition adaptation states of melanoma. Cell Discov 2016; 2:16028. [PMID: 27648299 PMCID: PMC5012007 DOI: 10.1038/celldisc.2016.28] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2015] [Accepted: 06/26/2016] [Indexed: 12/26/2022] Open
Abstract
A prominent mechanism of acquired resistance to BRAF inhibitors in BRAF (V600) -mutant melanoma is associated with the upregulation of receptor tyrosine kinases. Evidences suggested that this resistance mechanism is part of a more complex cellular adaptation process. Using an integrative strategy, we found this mechanism to invoke extensive transcriptomic, (phospho-) proteomic and phenotypic alterations that accompany a cellular transition to a de-differentiated, mesenchymal and invasive state. Even short-term BRAF-inhibitor exposure leads to an early adaptive, differentiation state change-characterized by a slow-cycling, persistent state. The early persistent state is distinct from the late proliferative, resistant state. However, both differentiation states share common signaling alterations including JUN upregulation. Motivated by the similarities, we found that co-targeting of BRAF and JUN is synergistic in killing fully resistant cells; and when used up-front, co-targeting substantially impairs the formation of the persistent subpopulation. We confirmed that JUN upregulation is a common response to BRAF inhibitor treatment in clinically treated patient tumors. Our findings demonstrate that events shared between early- and late-adaptation states provide candidate up-front co-treatment targets.
Collapse
|
221
|
Diamantopoulos P, Gogas H. Melanoma immunotherapy dominates the field. ANNALS OF TRANSLATIONAL MEDICINE 2016; 4:269. [PMID: 27563656 DOI: 10.21037/atm.2016.06.32] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The incidence of melanoma is increasing worldwide and despite early detection and intervention, the number of patients dying from metastatic disease continues to rise. The prognosis of advanced melanoma remains poor, with median survival between 6 and 9 months. Over the past 30 years and despite extensive clinical research, the treatment options for metastatic disease were limited and melanoma is still considered as one of the most therapy-resistant malignancies. Single-agent and combination chemotherapy, hormonal therapy, biochemotherapy, immunotherapy, targeted agent therapy and combination regimens failed to show a significant improvement in overall survival (OS). Recent advances and in-depth understanding of the biology of melanoma, have contributed to the development of new agents. Based on the molecular and immunological background of the disease, these new drugs have shown benefit in overall and progression-free survival (PFS). As the picture of the disease begins to change, oncologists need to alter their approach to melanoma treatment and consider disease biology together with targeted individualized treatment. In this review the authors attempt to offer an insight in the present and past melanoma treatment options, with a focus on the recently approved immunotherapeutic agents and the clinical perspectives of these new weapons against metastatic melanoma.
Collapse
Affiliation(s)
- Panagiotis Diamantopoulos
- 1 Department of Medicine, Laiko Hospital, National and Kapodistrian University of Athens School of Medicine, Athens, Greece
| | - Helen Gogas
- 1 Department of Medicine, Laiko Hospital, National and Kapodistrian University of Athens School of Medicine, Athens, Greece
| |
Collapse
|
222
|
Longoria TC, Tewari KS. Evaluation of the pharmacokinetics and metabolism of pembrolizumab in the treatment of melanoma. Expert Opin Drug Metab Toxicol 2016; 12:1247-53. [PMID: 27485741 DOI: 10.1080/17425255.2016.1216976] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Advanced melanoma is a devastating disease that has propelled research in therapeutics beyond chemotherapy and radiotherapy. Being highly immunogenic, melanoma is a model tumor for immunotherapy and has highlighted the therapeutic potential of the immune checkpoint inhibitors. AREAS COVERED This review discusses the pharmacologic properties, clinical efficacy, and safety profile of pembrolizumab, an IgG4-kappa humanized monoclonal antibody against the programmed cell death protein 1 (PD-1) receptor, for the treatment of unresectable or metastatic melanoma. EXPERT OPINION Pembrolizumab was the first PD-1 inhibitor to be approved by the U.S. Food and Drug Administration (FDA). Remarkably, this accelerated approval for the treatment of advanced, heavily pretreated melanoma was based on response rates alone from a phase I trial. As anticipated, pembrolizumab confirmed a survival advantage in phase II and III trials and has led the way for the study of other drugs that share its mechanism of action. Defining disease and patient characteristics associated with a response remains amongst the most pressing priorities.
Collapse
Affiliation(s)
- Teresa C Longoria
- a University of California , Irvine Medical Center , Orange , CA , USA
| | | |
Collapse
|
223
|
Fattore L, Malpicci D, Marra E, Belleudi F, Noto A, De Vitis C, Pisanu ME, Coluccia P, Camerlingo R, Roscilli G, Ribas A, Di Napoli A, Torrisi MR, Aurisicchio L, Ascierto PA, Mancini R, Ciliberto G. Combination of antibodies directed against different ErbB3 surface epitopes prevents the establishment of resistance to BRAF/MEK inhibitors in melanoma. Oncotarget 2016. [PMID: 26208478 PMCID: PMC4694796 DOI: 10.18632/oncotarget.4485] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Patients with metastatic melanoma bearing V600 mutations in BRAF oncogene clinically benefit from the treatment with BRAF inhibitors alone or in combination with MEK inhibitors. However, a limitation to such treatment is the occurrence of resistance. Tackling the adaptive changes helping cells survive from drug treatment may offer new therapeutic opportunities. Very recently the ErbB3 receptor has been shown to act as a central node promoting survival of BRAF mutated melanoma. In this paper we first demonstrate that ErbB3/AKT hyperphosphorylation occurs in BRAF mutated melanoma cell lines following exposure to BRAF and/or MEK inhibitors. This strongly correlates with increased transcriptional activation of its ligand neuregulin. Anti-ErbB3 antibodies impair the establishment of de novo cell resistance to BRAF inhibition in vitro. In order to more potently ablate ErbB3 activity we used a combination of two anti-ErbB3 antibodies directed against distinct epitopes of its extracellular domain. These two antibodies in combo with BRAF/MEK inhibitors potently inhibit in vitro cell growth and tumor regrowth after drug withdrawal in an in vivo xenograft model. Importantly, residual tumor masses from mice treated by the antibodies and BRAF/ERK inhibitors combo are characterized almost exclusively by large necrotic areas with limited residual areas of tumor growth. Taken together, our findings support the concept that triple therapy directed against BRAF/MEK/ErbB3 may be able to provide durable control of BRAF mutated metastatic melanoma.
Collapse
Affiliation(s)
- Luigi Fattore
- Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione G. Pascale", Naples, Italy.,Dipartimento di Chirurgia "P. Valdoni", Sapienza Università di Roma, Rome, Italy
| | - Debora Malpicci
- Dipartimento di Chirurgia "P. Valdoni", Sapienza Università di Roma, Rome, Italy.,Dipartimento di Medicina Sperimentale e Clinica, Università degli Studi di Catanzaro "Magna Graecia", Catanzaro, Italy
| | | | - Francesca Belleudi
- Dipartimento di Medicina Clinica e Molecolare, Sapienza Università di Roma, Rome, Italy.,Istituto Pasteur Fondazione Cenci Bolognetti, Dipartimento di Medicina Clinica e Molecolare, Sapienza Università di Roma, Rome, Italy
| | - Alessia Noto
- Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione G. Pascale", Naples, Italy.,Dipartimento di Chirurgia "P. Valdoni", Sapienza Università di Roma, Rome, Italy
| | - Claudia De Vitis
- Dipartimento di Chirurgia "P. Valdoni", Sapienza Università di Roma, Rome, Italy.,Dipartimento di Medicina Clinica e Molecolare, Sapienza Università di Roma, Rome, Italy
| | - Maria Elena Pisanu
- Dipartimento di Chirurgia "P. Valdoni", Sapienza Università di Roma, Rome, Italy.,Dipartimento di Medicina Sperimentale e Clinica, Università degli Studi di Catanzaro "Magna Graecia", Catanzaro, Italy
| | - Pierpaolo Coluccia
- Dipartimento di Chirurgia "P. Valdoni", Sapienza Università di Roma, Rome, Italy
| | - Rosa Camerlingo
- Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione G. Pascale", Naples, Italy
| | - Giuseppe Roscilli
- Takis S.r.l., Rome, Italy.,Dipartimento di Medicina Clinica e Molecolare, Sapienza Università di Roma, Rome, Italy
| | - Antoni Ribas
- Department of Medicine, Division of Hematology/Oncology, University of California Los Angeles (UCLA), Los Angeles, CA, USA
| | - Arianna Di Napoli
- Dipartimento di Medicina Clinica e Molecolare, Sapienza Università di Roma, Rome, Italy.,Azienda Ospedaliera S. Andrea, Rome, Italy
| | - Maria Rosaria Torrisi
- Dipartimento di Medicina Clinica e Molecolare, Sapienza Università di Roma, Rome, Italy.,Istituto Pasteur Fondazione Cenci Bolognetti, Dipartimento di Medicina Clinica e Molecolare, Sapienza Università di Roma, Rome, Italy.,Azienda Ospedaliera S. Andrea, Rome, Italy
| | | | - Paolo Antonio Ascierto
- Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione G. Pascale", Naples, Italy
| | - Rita Mancini
- Dipartimento di Chirurgia "P. Valdoni", Sapienza Università di Roma, Rome, Italy.,Dipartimento di Medicina Clinica e Molecolare, Sapienza Università di Roma, Rome, Italy
| | - Gennaro Ciliberto
- Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione G. Pascale", Naples, Italy
| |
Collapse
|
224
|
Combined vemurafenib and fotemustine in patients with BRAF V600 melanoma progressing on vemurafenib. Oncotarget 2016; 9:12408-12417. [PMID: 29552321 PMCID: PMC5844757 DOI: 10.18632/oncotarget.10589] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 06/30/2016] [Indexed: 01/08/2023] Open
Abstract
Background BRAF inhibitor vemurafenib achieves high response rate and an improvement in survival in patients with BRAF-mutated metastatic melanoma. However, median progression-free survival is only 6.9 months in the phase 3 study. Retrospective analyses suggest that treatment with BRAF inhibitors beyond initial progression might be associated with improved overall survival. We aimed to prospectively investigate the activity of prolonged treatment with vemurafenib and the addition of fotemustine in patients with systemic progression on prior single-agent BRAF inhibitor. Patients and Methods In this two-centres, single-arm Phase 2 trial, we enrolled patients with systemic progressive disease during single-agent vemurafenib treatment. Participants received vemurafenib 960 mg twice daily or dose administered at time of disease progression with vemurafenib previous treatment and fotemustine 100 mg/m2 intravenously every three weeks. The primary endpoint was PFS. Results Thirty-one patients were enrolled in the study; 16 patients had brain metastases at baseline. Median PFS was 3.9 months and 19 patients (61.3%) achieved disease control (1 CR, 4 PR, 14 SD). For patients achieving disease control, median duration of treatment was 6 months. Median OS was 5.8 months from enrolment and 15.4 months from start of previous vemurafenib. Five patients (16.1%) had a G3-4 AE, the most common being thrombocytopenia, which occurred in 3 patients. This trial is registered with ClinicalTrials.gov number NCT01983124. Conclusion The combination of vemurafenib plus fotemustine has clinical activity and an acceptable safety profile in BRAF-refractory patients.
Collapse
|
225
|
Rapid progression of intracranial melanoma metastases controlled with combined BRAF/MEK inhibition after discontinuation of therapy: a clinical challenge. J Neurooncol 2016; 129:389-393. [DOI: 10.1007/s11060-016-2196-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 07/03/2016] [Indexed: 11/26/2022]
|
226
|
Lagmay JP, Krailo MD, Dang H, Kim A, Hawkins DS, Beaty O, Widemann BC, Zwerdling T, Bomgaars L, Langevin AM, Grier HE, Weigel B, Blaney SM, Gorlick R, Janeway KA. Outcome of Patients With Recurrent Osteosarcoma Enrolled in Seven Phase II Trials Through Children's Cancer Group, Pediatric Oncology Group, and Children's Oncology Group: Learning From the Past to Move Forward. J Clin Oncol 2016; 34:3031-8. [PMID: 27400942 DOI: 10.1200/jco.2015.65.5381] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
PURPOSE The use of radiographic response as the primary end point in phase II osteosarcoma trials may limit optimal detection of treatment response because of the calcified tumor matrix. We performed this study to determine if time to progression could be used as an end point for subsequent studies. PATIENTS AND METHODS We performed a retrospective analysis of outcome for patients with recurrent/refractory osteosarcoma enrolled in one of seven phase II trials conducted by the Children's Oncology Group and predecessor groups from 1997 to 2007. All trials used RECIST or WHO radiographic response criteria and the primary end point of response rate. The following potential prognostic factors-age, trial, number of prior chemotherapy regimens, sex, and race/ethnicity-were evaluated for their impact on event-free survival (EFS). We used data from a phase II study (AOST0221) of patients with osteosarcoma who were given inhaled granulocyte-macrophage colony-stimulating factor with first pulmonary recurrence who had an EFS as well as biologic end point to determine the historical disease control rate for patients with fully resected disease. RESULTS In each included trial, the drugs tested were determined to be inactive on the basis of radiographic response rates. The EFS for 96 patients with osteosarcoma and measurable disease was 12% at 4 months (95% CI, 6% to 19%). There was no significant difference in EFS across trials according to number of prior treatment regimens or patient age, sex, and ethnicity. The 12-month EFS for the 42 evaluable patients enrolled in AOST0221 was 20% (95% CI, 10% to 34%). CONCLUSION The EFS was uniformly poor for children with recurrent/refractory osteosarcoma in these single-arm phase II trials. We have now constructed baseline EFS outcomes that can be used as a comparison for future phase II trials for recurrent osteosarcoma.
Collapse
Affiliation(s)
- Joanne P Lagmay
- Joanne P. Lagmay, Shands Hospital for Children, University of Florida, Gainesville, FL; Mark D. Krailo and Ha Dang, University of Southern California, Los Angeles; and Children's Oncology Group, Monrovia; Theodore Zwerdling, Jonathan Jaques Children's Cancer Center, Miller Children's and Women's Hospital, Long Beach, CA; AeRang Kim, Center for Cancer and Blood Disorders, Children's National Medical Center, Washington, DC; Douglas S. Hawkins, Seattle Children's Hospital, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA; Orren Beaty III, Zeiss Children's Cancer Center, Mission Hospitals, Asheville, NC; Brigitte C. Widemann, National Institutes of Health Clinical Center, Bethesda, MD; Lisa Bomgaars and Susan M. Blaney, Baylor College of Medicine/Texas Children's Cancer Center, Houston; Anne-Marie Langevin, University of Texas Health Science Center at San Antonio, San Antonio, TX; Holcombe E. Grier and Katherine A. Janeway, Dana-Farber Cancer Institute, Boston Children's Cancer and Blood Disorders Center, Boston, MA; Brenda Weigel, University of Minnesota, Minneapolis, MN; and Richard Gorlick, The Albert Einstein College of Medicine of Yeshiva University, The Children's Hospital at Montefiore, New York, NY.
| | - Mark D Krailo
- Joanne P. Lagmay, Shands Hospital for Children, University of Florida, Gainesville, FL; Mark D. Krailo and Ha Dang, University of Southern California, Los Angeles; and Children's Oncology Group, Monrovia; Theodore Zwerdling, Jonathan Jaques Children's Cancer Center, Miller Children's and Women's Hospital, Long Beach, CA; AeRang Kim, Center for Cancer and Blood Disorders, Children's National Medical Center, Washington, DC; Douglas S. Hawkins, Seattle Children's Hospital, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA; Orren Beaty III, Zeiss Children's Cancer Center, Mission Hospitals, Asheville, NC; Brigitte C. Widemann, National Institutes of Health Clinical Center, Bethesda, MD; Lisa Bomgaars and Susan M. Blaney, Baylor College of Medicine/Texas Children's Cancer Center, Houston; Anne-Marie Langevin, University of Texas Health Science Center at San Antonio, San Antonio, TX; Holcombe E. Grier and Katherine A. Janeway, Dana-Farber Cancer Institute, Boston Children's Cancer and Blood Disorders Center, Boston, MA; Brenda Weigel, University of Minnesota, Minneapolis, MN; and Richard Gorlick, The Albert Einstein College of Medicine of Yeshiva University, The Children's Hospital at Montefiore, New York, NY
| | - Ha Dang
- Joanne P. Lagmay, Shands Hospital for Children, University of Florida, Gainesville, FL; Mark D. Krailo and Ha Dang, University of Southern California, Los Angeles; and Children's Oncology Group, Monrovia; Theodore Zwerdling, Jonathan Jaques Children's Cancer Center, Miller Children's and Women's Hospital, Long Beach, CA; AeRang Kim, Center for Cancer and Blood Disorders, Children's National Medical Center, Washington, DC; Douglas S. Hawkins, Seattle Children's Hospital, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA; Orren Beaty III, Zeiss Children's Cancer Center, Mission Hospitals, Asheville, NC; Brigitte C. Widemann, National Institutes of Health Clinical Center, Bethesda, MD; Lisa Bomgaars and Susan M. Blaney, Baylor College of Medicine/Texas Children's Cancer Center, Houston; Anne-Marie Langevin, University of Texas Health Science Center at San Antonio, San Antonio, TX; Holcombe E. Grier and Katherine A. Janeway, Dana-Farber Cancer Institute, Boston Children's Cancer and Blood Disorders Center, Boston, MA; Brenda Weigel, University of Minnesota, Minneapolis, MN; and Richard Gorlick, The Albert Einstein College of Medicine of Yeshiva University, The Children's Hospital at Montefiore, New York, NY
| | - AeRang Kim
- Joanne P. Lagmay, Shands Hospital for Children, University of Florida, Gainesville, FL; Mark D. Krailo and Ha Dang, University of Southern California, Los Angeles; and Children's Oncology Group, Monrovia; Theodore Zwerdling, Jonathan Jaques Children's Cancer Center, Miller Children's and Women's Hospital, Long Beach, CA; AeRang Kim, Center for Cancer and Blood Disorders, Children's National Medical Center, Washington, DC; Douglas S. Hawkins, Seattle Children's Hospital, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA; Orren Beaty III, Zeiss Children's Cancer Center, Mission Hospitals, Asheville, NC; Brigitte C. Widemann, National Institutes of Health Clinical Center, Bethesda, MD; Lisa Bomgaars and Susan M. Blaney, Baylor College of Medicine/Texas Children's Cancer Center, Houston; Anne-Marie Langevin, University of Texas Health Science Center at San Antonio, San Antonio, TX; Holcombe E. Grier and Katherine A. Janeway, Dana-Farber Cancer Institute, Boston Children's Cancer and Blood Disorders Center, Boston, MA; Brenda Weigel, University of Minnesota, Minneapolis, MN; and Richard Gorlick, The Albert Einstein College of Medicine of Yeshiva University, The Children's Hospital at Montefiore, New York, NY
| | - Douglas S Hawkins
- Joanne P. Lagmay, Shands Hospital for Children, University of Florida, Gainesville, FL; Mark D. Krailo and Ha Dang, University of Southern California, Los Angeles; and Children's Oncology Group, Monrovia; Theodore Zwerdling, Jonathan Jaques Children's Cancer Center, Miller Children's and Women's Hospital, Long Beach, CA; AeRang Kim, Center for Cancer and Blood Disorders, Children's National Medical Center, Washington, DC; Douglas S. Hawkins, Seattle Children's Hospital, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA; Orren Beaty III, Zeiss Children's Cancer Center, Mission Hospitals, Asheville, NC; Brigitte C. Widemann, National Institutes of Health Clinical Center, Bethesda, MD; Lisa Bomgaars and Susan M. Blaney, Baylor College of Medicine/Texas Children's Cancer Center, Houston; Anne-Marie Langevin, University of Texas Health Science Center at San Antonio, San Antonio, TX; Holcombe E. Grier and Katherine A. Janeway, Dana-Farber Cancer Institute, Boston Children's Cancer and Blood Disorders Center, Boston, MA; Brenda Weigel, University of Minnesota, Minneapolis, MN; and Richard Gorlick, The Albert Einstein College of Medicine of Yeshiva University, The Children's Hospital at Montefiore, New York, NY
| | - Orren Beaty
- Joanne P. Lagmay, Shands Hospital for Children, University of Florida, Gainesville, FL; Mark D. Krailo and Ha Dang, University of Southern California, Los Angeles; and Children's Oncology Group, Monrovia; Theodore Zwerdling, Jonathan Jaques Children's Cancer Center, Miller Children's and Women's Hospital, Long Beach, CA; AeRang Kim, Center for Cancer and Blood Disorders, Children's National Medical Center, Washington, DC; Douglas S. Hawkins, Seattle Children's Hospital, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA; Orren Beaty III, Zeiss Children's Cancer Center, Mission Hospitals, Asheville, NC; Brigitte C. Widemann, National Institutes of Health Clinical Center, Bethesda, MD; Lisa Bomgaars and Susan M. Blaney, Baylor College of Medicine/Texas Children's Cancer Center, Houston; Anne-Marie Langevin, University of Texas Health Science Center at San Antonio, San Antonio, TX; Holcombe E. Grier and Katherine A. Janeway, Dana-Farber Cancer Institute, Boston Children's Cancer and Blood Disorders Center, Boston, MA; Brenda Weigel, University of Minnesota, Minneapolis, MN; and Richard Gorlick, The Albert Einstein College of Medicine of Yeshiva University, The Children's Hospital at Montefiore, New York, NY
| | - Brigitte C Widemann
- Joanne P. Lagmay, Shands Hospital for Children, University of Florida, Gainesville, FL; Mark D. Krailo and Ha Dang, University of Southern California, Los Angeles; and Children's Oncology Group, Monrovia; Theodore Zwerdling, Jonathan Jaques Children's Cancer Center, Miller Children's and Women's Hospital, Long Beach, CA; AeRang Kim, Center for Cancer and Blood Disorders, Children's National Medical Center, Washington, DC; Douglas S. Hawkins, Seattle Children's Hospital, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA; Orren Beaty III, Zeiss Children's Cancer Center, Mission Hospitals, Asheville, NC; Brigitte C. Widemann, National Institutes of Health Clinical Center, Bethesda, MD; Lisa Bomgaars and Susan M. Blaney, Baylor College of Medicine/Texas Children's Cancer Center, Houston; Anne-Marie Langevin, University of Texas Health Science Center at San Antonio, San Antonio, TX; Holcombe E. Grier and Katherine A. Janeway, Dana-Farber Cancer Institute, Boston Children's Cancer and Blood Disorders Center, Boston, MA; Brenda Weigel, University of Minnesota, Minneapolis, MN; and Richard Gorlick, The Albert Einstein College of Medicine of Yeshiva University, The Children's Hospital at Montefiore, New York, NY
| | - Theodore Zwerdling
- Joanne P. Lagmay, Shands Hospital for Children, University of Florida, Gainesville, FL; Mark D. Krailo and Ha Dang, University of Southern California, Los Angeles; and Children's Oncology Group, Monrovia; Theodore Zwerdling, Jonathan Jaques Children's Cancer Center, Miller Children's and Women's Hospital, Long Beach, CA; AeRang Kim, Center for Cancer and Blood Disorders, Children's National Medical Center, Washington, DC; Douglas S. Hawkins, Seattle Children's Hospital, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA; Orren Beaty III, Zeiss Children's Cancer Center, Mission Hospitals, Asheville, NC; Brigitte C. Widemann, National Institutes of Health Clinical Center, Bethesda, MD; Lisa Bomgaars and Susan M. Blaney, Baylor College of Medicine/Texas Children's Cancer Center, Houston; Anne-Marie Langevin, University of Texas Health Science Center at San Antonio, San Antonio, TX; Holcombe E. Grier and Katherine A. Janeway, Dana-Farber Cancer Institute, Boston Children's Cancer and Blood Disorders Center, Boston, MA; Brenda Weigel, University of Minnesota, Minneapolis, MN; and Richard Gorlick, The Albert Einstein College of Medicine of Yeshiva University, The Children's Hospital at Montefiore, New York, NY
| | - Lisa Bomgaars
- Joanne P. Lagmay, Shands Hospital for Children, University of Florida, Gainesville, FL; Mark D. Krailo and Ha Dang, University of Southern California, Los Angeles; and Children's Oncology Group, Monrovia; Theodore Zwerdling, Jonathan Jaques Children's Cancer Center, Miller Children's and Women's Hospital, Long Beach, CA; AeRang Kim, Center for Cancer and Blood Disorders, Children's National Medical Center, Washington, DC; Douglas S. Hawkins, Seattle Children's Hospital, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA; Orren Beaty III, Zeiss Children's Cancer Center, Mission Hospitals, Asheville, NC; Brigitte C. Widemann, National Institutes of Health Clinical Center, Bethesda, MD; Lisa Bomgaars and Susan M. Blaney, Baylor College of Medicine/Texas Children's Cancer Center, Houston; Anne-Marie Langevin, University of Texas Health Science Center at San Antonio, San Antonio, TX; Holcombe E. Grier and Katherine A. Janeway, Dana-Farber Cancer Institute, Boston Children's Cancer and Blood Disorders Center, Boston, MA; Brenda Weigel, University of Minnesota, Minneapolis, MN; and Richard Gorlick, The Albert Einstein College of Medicine of Yeshiva University, The Children's Hospital at Montefiore, New York, NY
| | - Anne-Marie Langevin
- Joanne P. Lagmay, Shands Hospital for Children, University of Florida, Gainesville, FL; Mark D. Krailo and Ha Dang, University of Southern California, Los Angeles; and Children's Oncology Group, Monrovia; Theodore Zwerdling, Jonathan Jaques Children's Cancer Center, Miller Children's and Women's Hospital, Long Beach, CA; AeRang Kim, Center for Cancer and Blood Disorders, Children's National Medical Center, Washington, DC; Douglas S. Hawkins, Seattle Children's Hospital, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA; Orren Beaty III, Zeiss Children's Cancer Center, Mission Hospitals, Asheville, NC; Brigitte C. Widemann, National Institutes of Health Clinical Center, Bethesda, MD; Lisa Bomgaars and Susan M. Blaney, Baylor College of Medicine/Texas Children's Cancer Center, Houston; Anne-Marie Langevin, University of Texas Health Science Center at San Antonio, San Antonio, TX; Holcombe E. Grier and Katherine A. Janeway, Dana-Farber Cancer Institute, Boston Children's Cancer and Blood Disorders Center, Boston, MA; Brenda Weigel, University of Minnesota, Minneapolis, MN; and Richard Gorlick, The Albert Einstein College of Medicine of Yeshiva University, The Children's Hospital at Montefiore, New York, NY
| | - Holcombe E Grier
- Joanne P. Lagmay, Shands Hospital for Children, University of Florida, Gainesville, FL; Mark D. Krailo and Ha Dang, University of Southern California, Los Angeles; and Children's Oncology Group, Monrovia; Theodore Zwerdling, Jonathan Jaques Children's Cancer Center, Miller Children's and Women's Hospital, Long Beach, CA; AeRang Kim, Center for Cancer and Blood Disorders, Children's National Medical Center, Washington, DC; Douglas S. Hawkins, Seattle Children's Hospital, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA; Orren Beaty III, Zeiss Children's Cancer Center, Mission Hospitals, Asheville, NC; Brigitte C. Widemann, National Institutes of Health Clinical Center, Bethesda, MD; Lisa Bomgaars and Susan M. Blaney, Baylor College of Medicine/Texas Children's Cancer Center, Houston; Anne-Marie Langevin, University of Texas Health Science Center at San Antonio, San Antonio, TX; Holcombe E. Grier and Katherine A. Janeway, Dana-Farber Cancer Institute, Boston Children's Cancer and Blood Disorders Center, Boston, MA; Brenda Weigel, University of Minnesota, Minneapolis, MN; and Richard Gorlick, The Albert Einstein College of Medicine of Yeshiva University, The Children's Hospital at Montefiore, New York, NY
| | - Brenda Weigel
- Joanne P. Lagmay, Shands Hospital for Children, University of Florida, Gainesville, FL; Mark D. Krailo and Ha Dang, University of Southern California, Los Angeles; and Children's Oncology Group, Monrovia; Theodore Zwerdling, Jonathan Jaques Children's Cancer Center, Miller Children's and Women's Hospital, Long Beach, CA; AeRang Kim, Center for Cancer and Blood Disorders, Children's National Medical Center, Washington, DC; Douglas S. Hawkins, Seattle Children's Hospital, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA; Orren Beaty III, Zeiss Children's Cancer Center, Mission Hospitals, Asheville, NC; Brigitte C. Widemann, National Institutes of Health Clinical Center, Bethesda, MD; Lisa Bomgaars and Susan M. Blaney, Baylor College of Medicine/Texas Children's Cancer Center, Houston; Anne-Marie Langevin, University of Texas Health Science Center at San Antonio, San Antonio, TX; Holcombe E. Grier and Katherine A. Janeway, Dana-Farber Cancer Institute, Boston Children's Cancer and Blood Disorders Center, Boston, MA; Brenda Weigel, University of Minnesota, Minneapolis, MN; and Richard Gorlick, The Albert Einstein College of Medicine of Yeshiva University, The Children's Hospital at Montefiore, New York, NY
| | - Susan M Blaney
- Joanne P. Lagmay, Shands Hospital for Children, University of Florida, Gainesville, FL; Mark D. Krailo and Ha Dang, University of Southern California, Los Angeles; and Children's Oncology Group, Monrovia; Theodore Zwerdling, Jonathan Jaques Children's Cancer Center, Miller Children's and Women's Hospital, Long Beach, CA; AeRang Kim, Center for Cancer and Blood Disorders, Children's National Medical Center, Washington, DC; Douglas S. Hawkins, Seattle Children's Hospital, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA; Orren Beaty III, Zeiss Children's Cancer Center, Mission Hospitals, Asheville, NC; Brigitte C. Widemann, National Institutes of Health Clinical Center, Bethesda, MD; Lisa Bomgaars and Susan M. Blaney, Baylor College of Medicine/Texas Children's Cancer Center, Houston; Anne-Marie Langevin, University of Texas Health Science Center at San Antonio, San Antonio, TX; Holcombe E. Grier and Katherine A. Janeway, Dana-Farber Cancer Institute, Boston Children's Cancer and Blood Disorders Center, Boston, MA; Brenda Weigel, University of Minnesota, Minneapolis, MN; and Richard Gorlick, The Albert Einstein College of Medicine of Yeshiva University, The Children's Hospital at Montefiore, New York, NY
| | - Richard Gorlick
- Joanne P. Lagmay, Shands Hospital for Children, University of Florida, Gainesville, FL; Mark D. Krailo and Ha Dang, University of Southern California, Los Angeles; and Children's Oncology Group, Monrovia; Theodore Zwerdling, Jonathan Jaques Children's Cancer Center, Miller Children's and Women's Hospital, Long Beach, CA; AeRang Kim, Center for Cancer and Blood Disorders, Children's National Medical Center, Washington, DC; Douglas S. Hawkins, Seattle Children's Hospital, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA; Orren Beaty III, Zeiss Children's Cancer Center, Mission Hospitals, Asheville, NC; Brigitte C. Widemann, National Institutes of Health Clinical Center, Bethesda, MD; Lisa Bomgaars and Susan M. Blaney, Baylor College of Medicine/Texas Children's Cancer Center, Houston; Anne-Marie Langevin, University of Texas Health Science Center at San Antonio, San Antonio, TX; Holcombe E. Grier and Katherine A. Janeway, Dana-Farber Cancer Institute, Boston Children's Cancer and Blood Disorders Center, Boston, MA; Brenda Weigel, University of Minnesota, Minneapolis, MN; and Richard Gorlick, The Albert Einstein College of Medicine of Yeshiva University, The Children's Hospital at Montefiore, New York, NY
| | - Katherine A Janeway
- Joanne P. Lagmay, Shands Hospital for Children, University of Florida, Gainesville, FL; Mark D. Krailo and Ha Dang, University of Southern California, Los Angeles; and Children's Oncology Group, Monrovia; Theodore Zwerdling, Jonathan Jaques Children's Cancer Center, Miller Children's and Women's Hospital, Long Beach, CA; AeRang Kim, Center for Cancer and Blood Disorders, Children's National Medical Center, Washington, DC; Douglas S. Hawkins, Seattle Children's Hospital, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA; Orren Beaty III, Zeiss Children's Cancer Center, Mission Hospitals, Asheville, NC; Brigitte C. Widemann, National Institutes of Health Clinical Center, Bethesda, MD; Lisa Bomgaars and Susan M. Blaney, Baylor College of Medicine/Texas Children's Cancer Center, Houston; Anne-Marie Langevin, University of Texas Health Science Center at San Antonio, San Antonio, TX; Holcombe E. Grier and Katherine A. Janeway, Dana-Farber Cancer Institute, Boston Children's Cancer and Blood Disorders Center, Boston, MA; Brenda Weigel, University of Minnesota, Minneapolis, MN; and Richard Gorlick, The Albert Einstein College of Medicine of Yeshiva University, The Children's Hospital at Montefiore, New York, NY
| |
Collapse
|
227
|
Kim A, Cohen MS. The discovery of vemurafenib for the treatment of BRAF-mutated metastatic melanoma. Expert Opin Drug Discov 2016; 11:907-16. [PMID: 27327499 DOI: 10.1080/17460441.2016.1201057] [Citation(s) in RCA: 103] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
INTRODUCTION In the era of precision medicine and sophisticated modern genetics, the discovery of the BRAF(V600) inhibitor, vemurafenib, quickly became the model for targeted therapy in melanomas. As early as 2002, the majority of metastatic melanomas were described to harbor the BRAF(V600) mutation, setting the stage for an explosion of interest for targeting this protein as a novel therapeutic strategy. The highly selective BRAF(V600) inhibitor, vemurafenib, was identified initially through a large-scale drug screen. AREAS COVERED Here we examine vemurafenib's journey from discovery to clinical use in metastatic melanoma. Topics covered include preclinical data, single agent Phase 1,2 and 3 clinical trials, resistance issues and mechanisms, adverse effects including the development of squamous cell cancers, and combination trials. EXPERT OPINION Due to its tolerance, low toxicity profile, rapid tumor response, and improved outcomes in melanoma patients with BRAF(V600) mutations, vemurafenib was advanced rapidly through clinical trials to receive FDA approval in 2011. While its efficacy is well documented, durability has become an issue for most patients who experience therapeutic resistance in approximately 6-8 months. In addition, a concerning toxicity observed in patients taking the drug include development of localized cutaneous squamous cell carcinomas (SCCs). It is hypothesized that drug resistance and SCC development result from a similar paradoxical activation of protein signaling pathways, specifically MAPK. Identification of these mechanisms has led to additional treatment strategies involving new combination therapies.
Collapse
Affiliation(s)
- Alex Kim
- a Department of Surgery , University of Michigan , Ann Arbor , MI , USA
| | - Mark S Cohen
- a Department of Surgery , University of Michigan , Ann Arbor , MI , USA
| |
Collapse
|
228
|
Filippi AR, Fava P, Badellino S, Astrua C, Ricardi U, Quaglino P. Radiotherapy and immune checkpoints inhibitors for advanced melanoma. Radiother Oncol 2016; 120:1-12. [PMID: 27345592 DOI: 10.1016/j.radonc.2016.06.003] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 05/26/2016] [Accepted: 06/08/2016] [Indexed: 01/05/2023]
Abstract
INTRODUCTION The therapeutic landscape of metastatic melanoma drastically changed after the introduction of targeted therapies and immunotherapy, in particular immune checkpoints inhibitors (ICI). In recent years, positive effects on the immune system associated to radiotherapy (RT) were discovered, and radiation has been tested in combination with ICI in both pre-clinical and clinical studies (many of them still ongoing). We here summarize the rationale and the preliminary clinical results of this approach. MATERIALS AND METHODS In the first part of this review article, redacted with narrative non-systematic methodology, we describe the clinical results of immune checkpoints blockade in melanoma as well as the biological basis for the combination of ICI with RT; in the second part, we systematically review scientific publications reporting on the clinical results of the combination of ICI and RT for advanced melanoma. RESULTS The biological and mechanistic rationale behind the combination of ICI and radiation is well supported by several preclinical findings. Retrospective observational series and few prospective trials support the potential synergistic effect between radiation and ICI for metastatic melanoma. CONCLUSION RT may potentiate anti-melanoma activity of ICI by enhancing response on both target and non-target lesions. Several prospective trials are ongoing with the aim of further exploring this combination in the clinical setting, hopefully confirming initial observations and opening a new therapeutic window for advanced melanoma patients.
Collapse
Affiliation(s)
| | - Paolo Fava
- Department of Medical Sciences, Dermatology/Oncology, University of Torino, Italy
| | - Serena Badellino
- Department of Oncology, Radiation Oncology, University of Torino, Italy
| | - Chiara Astrua
- Department of Medical Sciences, Dermatology/Oncology, University of Torino, Italy
| | - Umberto Ricardi
- Department of Oncology, Radiation Oncology, University of Torino, Italy
| | - Pietro Quaglino
- Department of Medical Sciences, Dermatology/Oncology, University of Torino, Italy
| |
Collapse
|
229
|
Progression-free survival landmark analysis: a critical endpoint in melanoma clinical trials. Lancet Oncol 2016; 17:1037-1039. [PMID: 27324281 DOI: 10.1016/s1470-2045(16)30017-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2016] [Revised: 03/08/2016] [Accepted: 03/15/2016] [Indexed: 11/20/2022]
|
230
|
Wang C, Xu W, Hao W, Wang B, Zheng Q. Alternol inhibits the proliferation and induces the differentiation of the mouse melanoma B16F0 cell line. Oncol Rep 2016; 36:1150-6. [PMID: 27278753 DOI: 10.3892/or.2016.4844] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2015] [Accepted: 03/02/2016] [Indexed: 11/06/2022] Open
Abstract
High malignant potential and low susceptibility to treatment are characteristics of malignant melanoma. Alternol, a novel compound purified from microbial fermentation products obtained from the bark of the yew tree, exhibits a variety of antitumor activities. Based on these findings, the aim of the present study was to extend the knowledge on the antineoplastic effect of alternol in the mouse melanoma B16F0 cell line. Alternol significantly inhibited the proliferation and colony formation of B16F0 cells in a dose-dependent manner as detected by MTT and soft agar colony formation assays. NaOH alkaline lysis and oxidation of Dopa indicated that alternol enhanced the melanin content and tyrosinase activity of the B16F0 cells and results also showed a dose‑response relationship. Morphologic changes accompanied by extended dendrites were discovered in the B16F0 cells after treatment with alternol. Furthermore, the mRNA levels of tyrosinase, Trp1 and Trp2 were increased by alternol. Our results confirmed that alternol possesses marked antineoplastic properties against melanoma cells, indicating that this microbial fermentation product is a promising agent for the differentiation therapy of cancer. The inhibition of cell proliferation and colony formation by alternol was associated with both cytotoxicity and induction of differentiation.
Collapse
Affiliation(s)
- Caixia Wang
- Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Wenjuan Xu
- Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Wenjin Hao
- Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Bingsheng Wang
- Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Qiusheng Zheng
- Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| |
Collapse
|
231
|
Petrelli F, Coinu A, Cabiddu M, Borgonovo K, Ghilardi M, Lonati V, Barni S. Early analysis of surrogate endpoints for metastatic melanoma in immune checkpoint inhibitor trials. Medicine (Baltimore) 2016; 95:e3997. [PMID: 27368007 PMCID: PMC4937921 DOI: 10.1097/md.0000000000003997] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Recent major phase III trials led to the approval of immune checkpoint inhibitors (ipilimumab, pembrolizumab, and nivolumab) in metastatic malignant melanoma (MM). We aim to assess whether median progression-free survival, and 1 and 2-year overall survival (OS) rates are reliable surrogate endpoints for median OS through a meta-analysis of published trials involving immunotherapy. A systematic literature search in PubMed, EMBASE, Web of Science, and SCOPUS of published phase II to III trials with immunotherapy as the treatment for MM was conducted. Adjusted weighted linear regression was used to calculate Pearson correlations (R) between surrogates and median OS, and between treatment effects on surrogates and median OS. A total of 13 studies involving 3373 patients with MM were identified. The correlation of progression-free survival with OS was not significant (R = 0.45, P = .11). Conversely, the correlation between 1-year OS and median OS was very strong (R = 0.93, 95% confidence interval [CI] 0.84-0.96, P < .00001), as was the correlation between 2-year OS and OS (R = 0.79, 95% CI 0.51-0.91, P = .0001). The correlation between the treatment effects on 1-year OS and OS was also significant (R = -0.86, 95% CI -0.3 to 0.97, P = .01). Similar results were obtained for 2-year OS. According to the available study data, 1-year OS rate could be regarded as a potential surrogate for median OS in novel immunotherapy trials of metastatic MM. Waiting for ongoing studies (e.g., pembrolizumab), we suggest that this intermediate endpoint could be considered as a potential primary endpoint in future clinical trials.
Collapse
Affiliation(s)
- Fausto Petrelli
- Oncology Department, Oncology Division, ASST Bergamo Ovest, Treviglio (BG), Italy
- Correspondence: Fausto Petrelli, Oncology Department, Oncology Division, ASST Bergamo Ovest, Piazzale Ospedale 1, 24047 Treviglio (BG), Italy (e-mail: )
| | | | | | | | | | | | | |
Collapse
|
232
|
Loo K, Daud A. Emerging biomarkers as predictors to anti-PD1/PD-L1 therapies in advanced melanoma. Immunotherapy 2016; 8:775-84. [DOI: 10.2217/imt-2016-0039] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Recent advances in the field of cancer immunotherapy have resulted in a surge of new therapies for patients spanning multiple cancer indications. In melanoma alone, several immunotherapies have emerged as promising agents to tackle the aggressive, often refractory disease in the advanced/metastatic setting. The Programmed Cell Death pathway, from which anti-PD-1 and anti-PD-L1 therapies were developed, has shown immense promise. Given the marked success of the PD-1/PD-L1 immunotherapies, several targets have emerged as promising biomarkers, including PD-L1 tumor expression, tumor-infiltrating T-cell markers, dendritic cell markers, TCR sequencing, neoantigens and peripheral blood markers. Highlighted in this review, we examine the recent efforts to identify robust and reliable biomarkers as predictors of response to anti-PD-1/PD-L1 immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Kimberly Loo
- Helen Diller Comprehensive Cancer Center, Department of Melanoma and Cutaneous Oncology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Adil Daud
- Helen Diller Comprehensive Cancer Center, Department of Melanoma and Cutaneous Oncology, University of California San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
233
|
Johnson DB, Pollack MH, Sosman JA. Emerging targeted therapies for melanoma. Expert Opin Emerg Drugs 2016; 21:195-207. [DOI: 10.1080/14728214.2016.1184644] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
234
|
Modelling Comparative Efficacy of Drugs with Different Survival Profiles: Ipilimumab, Vemurafenib and Dacarbazine in Advanced Melanoma. BioDrugs 2016; 30:307-19. [DOI: 10.1007/s40259-016-0178-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
235
|
Aya F, Fernandez-Martinez A, Gaba L, Victoria I, Tosca M, Pineda E, Gascon P, Prat A, Arance A. Sequential treatment with immunotherapy and BRAF inhibitors in BRAF-mutant advanced melanoma. Clin Transl Oncol 2016; 19:119-124. [PMID: 27147251 DOI: 10.1007/s12094-016-1514-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 04/18/2016] [Indexed: 10/21/2022]
Abstract
PURPOSE Immunotherapy (IT) agents and BRAF inhibitors (BRAFi) are effective treatments for patients with advanced BRAF-mutant melanoma although the optimal sequence remains to be elucidated. The aim of this study was to compare the outcomes of two different cohorts of patients treated with BRAFi first, then IT or the reverse sequence. PATIENTS AND METHODS This is a retrospective study on two groups of patients: a cohort was treated first with BRAFi followed by immunotherapy (BRAFi-IT) and the other cohort with the reverse sequence (IT-BRAFi). Baseline characteristics and clinical outcomes were compared between the two cohorts. RESULTS A total of 25 patients were included in the study. Sixteen patients were given BRAFi-IT sequence and nine received IT-BRAFi sequence. No differences were observed in the characteristics of patients prior to each treatment between cohorts. Objective response rate (ORR) achieved by BRAFi were not different among groups. ORR achieved by IT was higher when administered after BRAFi (43.8 vs 0 %). Survival rates at 1-2 years were similar in both cohorts and median overall survival was not different for BRAFi-IT and IT-BRAFi (log rank test p = 0.97). CONCLUSIONS No differences were observed in OS between the two cohorts. These results support the indistinct use of IT or BRAFi as initial treatment in patients with metastatic BRAF-mutant melanoma, although higher rate of response to IT was observed when administered after BRAFi. Prospective randomized clinical trials are needed on this issue.
Collapse
Affiliation(s)
- F Aya
- Department of Medical Oncology, Hospital Clínic, Barcelona, Spain. .,Translational Genomics and Targeted Therapeutics in Solid Tumors, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
| | - A Fernandez-Martinez
- Department of Medical Oncology, Hospital Clínic, Barcelona, Spain.,Translational Genomics and Targeted Therapeutics in Solid Tumors, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - L Gaba
- Department of Medical Oncology, Hospital Clínic, Barcelona, Spain.,Translational Genomics and Targeted Therapeutics in Solid Tumors, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - I Victoria
- Department of Medical Oncology, Hospital Clínic, Barcelona, Spain.,Translational Genomics and Targeted Therapeutics in Solid Tumors, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - M Tosca
- Department of Medical Oncology, Hospital Clínic, Barcelona, Spain.,Translational Genomics and Targeted Therapeutics in Solid Tumors, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - E Pineda
- Department of Medical Oncology, Hospital Clínic, Barcelona, Spain.,Translational Genomics and Targeted Therapeutics in Solid Tumors, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - P Gascon
- Department of Medical Oncology, Hospital Clínic, Barcelona, Spain
| | - A Prat
- Department of Medical Oncology, Hospital Clínic, Barcelona, Spain.,Translational Genomics and Targeted Therapeutics in Solid Tumors, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - A Arance
- Department of Medical Oncology, Hospital Clínic, Barcelona, Spain.,Translational Genomics and Targeted Therapeutics in Solid Tumors, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| |
Collapse
|
236
|
Abstract
The treatment of metastatic melanoma is rapidly changing. In 2002, the BRAF mutation was described in over 50% of melanomas and led to the first BRAF inhibitor, vemurafenib, being approved for clinical use in 2011. Clinical responses are often rapid but duration of response is limited due to the development of resistance. MEK is the next downstream target from BRAF in the MAP kinase pathway. Trametinib was the first MEK inhibitor to be approved for clinical use in 2013. Preclinical studies demonstrated a delay in resistance and a reduction in cutaneous toxicity by combined BRAF and MEK inhibition. Here, we review the rationale for clinical development of trametinib and give an update on recent clinical trials of trametinib alone and in combination with braf inhibition in melanoma.
Collapse
Affiliation(s)
- Neha Chopra
- Mount Vernon Hospital, Medical Oncology, Rickmansworth Road, Northwood, Middlesex, HA6 2RN, UK
| | | |
Collapse
|
237
|
Yao N, Jang S. Adoption of ipilimumab in the United States: a Medicare study. Expert Rev Pharmacoecon Outcomes Res 2016; 16:439-40. [PMID: 27097117 DOI: 10.1080/14737167.2016.1180979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Nengliang Yao
- a Department of Public Health Sciences , University of Virginia , Charlottesville , VA , USA
| | - Sekwon Jang
- b Inova Schar Cancer Institute, Melanoma and Skin Cancer Center , Fairfax , VA , USA
| |
Collapse
|
238
|
Liu JS, Rao S. Long-term drug costs per life-month gained associated with first-line treatments for unresectable or metastatic melanoma. Exp Hematol Oncol 2016; 5:9. [PMID: 27069772 PMCID: PMC4827222 DOI: 10.1186/s40164-016-0039-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 03/30/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND For unresectable or metastatic melanoma, first-line ipilimumab has demonstrated long-term survival benefits over a 7-year period. First-line treatment with BRAF inhibitors has demonstrated efficacy in clinical trials with up to 3 years of follow-up. The long-term comparative efficacy and costs of ipilimumab and BRAF inhibitors are unknown. METHODS Patient-level data from 12 clinical studies for ipilimumab were used. Survival data were extracted from included clinical trials for BRAF inhibitors based on a systematic literature review. Different parametric survival models, including exponential, Gompertz, log-normal, and Weibull models, were used to fit reported overall survival (OS) data and to project long-term survival for BRAF inhibitors. Survival benefits were measured in terms of total life-months gained as calculated by the area under the curve of OS Kaplan-Meier curves for the observed ipilimumab data and projected BRAF inhibitor data. Total life-months gained and cumulative costs per life-month gained were compared between ipilimumab and BRAF inhibitors. RESULTS The systematic literature review identified six randomized-controlled trials of BRAF inhibitors for subsequent analyses. With 7-year follow-up, ipilimumab was associated with a total of 28.5 life-months gained. Based on the Weibull model, the extrapolated total life-months gained for BRAF inhibitors were 26.5 months for dabrafenib, 21.3 months for trametinib, 14.3 months for vemurafenib, and 24.6 months for dabrafenib + trametinib. In sensitivity analyses, extrapolated total life-months gained varied across the three other models, ranging from 13.7 to 36.8 months across therapies. Cumulative costs per life-month gained with ipilimumab decreased steadily over time, while the costs remained constant for BRAF inhibitors due to continuous dosing. By year 3, cumulative costs per life-month gained were the lowest with ipilimumab; by year 7, the costs were $4281 for ipilimumab, compared with $8920 for dabrafenib, $10,211 for trametinib, $11,002 for vemurafenib, and $19,132 for the dabrafenib + trametinib combination therapy. CONCLUSIONS Ipilimumab was associated with a better long-term cost-per-life month compared to BRAF agents. Long-term extrapolation of survival with BRAF agents was uncertain, and showed no evidence of prolonged survival compared to ipilimumab.
Collapse
Affiliation(s)
- Jun S. Liu
- Department of Statistics, Harvard University, Cambridge, MA USA
| | | |
Collapse
|
239
|
Abstract
Recent data have demonstrated improved survival with targeted and immune therapies in patients with advanced melanoma, leading to much excitement amongst the oncology community and the widespread use of these drugs in combination regimens. However, the place of these combination therapies in the treatment of advanced melanoma remains to be fully determined. In this perspectives article, we critically review the available data and outline the rationale for these combinations being adopted as the standard of care for patients with advanced melanoma in the future.
Collapse
Affiliation(s)
- Keiran S M Smalley
- The Department of Cutaneous Oncology, Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL, USA.
- The Department of Tumor Biology, Moffitt Cancer Center and Research Institute, Tampa, FL, USA.
| | - Zeynep Eroglu
- The Department of Cutaneous Oncology, Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL, USA
| | - Vernon K Sondak
- The Department of Cutaneous Oncology, Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL, USA
| |
Collapse
|
240
|
Sullivan R. Serum lactate dehydrogenase is a more useful biomarker of prognosis than serum S100B in patients with BRAF-mutant melanoma. Br J Dermatol 2016; 174:716-7. [DOI: 10.1111/bjd.14503] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- R.J. Sullivan
- Center for Melanoma; Massachusetts General Hospital Cancer Center; 55 Fruit Street Boston MA 02114 U.S.A
| |
Collapse
|
241
|
Indirect Treatment Comparison of Talimogene Laherparepvec Compared with Ipilimumab and Vemurafenib for the Treatment of Patients with Metastatic Melanoma. Adv Ther 2016; 33:643-57. [PMID: 26979173 PMCID: PMC4846697 DOI: 10.1007/s12325-016-0313-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Indexed: 02/04/2023]
Abstract
INTRODUCTION Few randomized controlled trials have compared new treatments for metastatic melanoma. We sought to examine the relative treatment effect of talimogene laherparepvec compared with ipilimumab and vemurafenib. METHODS A systematic literature review of treatments for metastatic melanoma was undertaken but a valid network of evidence could not be established because of a lack of comparative data or studies with sufficient common comparators. A conventional adjusted indirect treatment comparison via network meta-analysis was, therefore, not feasible. Instead, a meta-analysis of absolute efficacy was undertaken, adjusting overall survival (OS) data for differences in prognostic factors between studies using a published algorithm. RESULTS Four trials were included in the final indirect treatment comparison: two of ipilimumab, one of vemurafenib, and one of talimogene laherparepvec. Median OS for ipilimumab and vemurafenib increased significantly when adjustment was applied, demonstrating that variation in disease and patient characteristics was biasing OS estimates; adjusting for this made the survival data more comparable. For both ipilimumab and vemurafenib, the adjustments improved Kaplan-Meier OS curves; the observed talimogene laherparepvec OS curve remained above the adjusted OS curves for ipilimumab and vemurafenib, showing that long-term survival could differ from the observed medians. CONCLUSION Even with limited data, talimogene laherparepvec, ipilimumab, and vemurafenib could be compared following adjustments, thereby providing a more reliable understanding of the relative effect of treatment on survival in a more comparable patient population. The results of this analysis suggest that OS with talimogene laherparepvec is at least as good as with ipilimumab and vemurafenib and improvement was more pronounced in patients with no bone, brain, lung or other visceral metastases. FUNDING Amgen Inc.
Collapse
|
242
|
Comin-Anduix B, Escuin-Ordinas H, Ibarrondo FJ. Tremelimumab: research and clinical development. Onco Targets Ther 2016; 9:1767-76. [PMID: 27042127 PMCID: PMC4809326 DOI: 10.2147/ott.s65802] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The immune checkpoint therapy is a relatively recent strategy that aims to tweak the immune system to effectively attack cancer cells. The understanding of the immune responses and their regulation at the intracellular level and the development of fully humanized monoclonal antibodies are the pillars of an approach that could elicit durable clinical responses and even remission in some patients with cancer. Most of the immune checkpoints that regulate the T-cell responses (activation and inhibition) operate through proteins present on the cytoplasmic membrane of the immune cells. Therefore, specific antibodies capable of blocking the inhibitory signals should lead to unrestrained immune responses that supersede the inhibitory mechanisms, which are naturally present in the tumor microenviroment. The best-known and most successful targets for immune checkpoint therapy are the cytotoxic T-lymphocyte antigen-4 and programmed cell death-1 coreceptors. Tremelimumab (CP-675,206) is a fully humanized monoclonal antibody specific for cytotoxic T-lymphocyte antigen-4, which has been successfully used to treat patients with metastatic melanoma and some other cancers. Although still a work in progress, the use of tremelimumab as an immune checkpoint therapeutic agent is a promising approach alone or in combination with other anticancer drugs. Here, we review the use of this antibody in a number of clinical trials against solid tumors.
Collapse
Affiliation(s)
- Begoña Comin-Anduix
- Division of Surgical-Oncology, Department of Surgery, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, USA; Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, USA
| | - Helena Escuin-Ordinas
- Division of Hematology-Oncology, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, USA
| | - Francisco Javier Ibarrondo
- Division of Infectious Diseases, Department of Medicine, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
243
|
Spagnolo F, Picasso V, Spano L, Tanda E, Venzano C, Queirolo P. Update on Metastatic Uveal Melanoma: Progress and Challenges. BioDrugs 2016; 30:161-72. [DOI: 10.1007/s40259-016-0167-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
244
|
Arance AM, Berrocal A, Lopez-Martin JA, de la Cruz-Merino L, Soriano V, Martín Algarra S, Alonso L, Cerezuela P, La Orden B, Espinosa E. Safety of vemurafenib in patients with BRAF V600 mutated metastatic melanoma: the Spanish experience. Clin Transl Oncol 2016; 18:1147-1157. [DOI: 10.1007/s12094-016-1498-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 03/02/2016] [Indexed: 10/22/2022]
|
245
|
Slimano F, Roessle C, Blanc C, De Maleissye MF, Bauler S. [Updates on prevention and treatment of melanoma: Pharmacist involvements and challenges]. ANNALES PHARMACEUTIQUES FRANÇAISES 2016; 74:335-49. [PMID: 26968263 DOI: 10.1016/j.pharma.2016.01.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 01/25/2016] [Accepted: 01/27/2016] [Indexed: 02/07/2023]
Abstract
Melanoma is a skin cancer that represents an actual public health problem. Its incidence is increasing every year. Environmental risk factors have been clearly identified. Early diagnosis of a suspicious skin lesion should be possible by any health professionals because the prognosis is correlated with the evolution of the disease and the presence of metastases. The advent of new therapies in metastatic forms with the development of immunotherapies and kinases inhibitors has significantly changed the management of this disease. New therapies are available in retail pharmacies and involve health professionals out of the hospital. This article is intended for community and hospital pharmacists and summarizes recommendations for primary and secondary prevention. It updates on new targeted therapies. It wants to give advices to the community pharmacists about the effective use of those treatments for melanoma.
Collapse
Affiliation(s)
- F Slimano
- Département de pharmacie clinique, Gustave-Roussy cancer campus, 114, rue Édouard-Vaillant, 94805 Villejuif, France; Laboratoire de pharmacologie et pharmacocinétique, UFR de pharmacie, université de Reims-Champagne-Ardenne, 51, rue Cognacq-Jay, 51100 Reims, France; Unité MEDyC, UMR CNRS/URCA, université de Reims-Champagne-Ardenne, 51, rue Cognacq-Jay, 51100 Reims, France.
| | - C Roessle
- Département de pharmacie clinique, Gustave-Roussy cancer campus, 114, rue Édouard-Vaillant, 94805 Villejuif, France
| | - C Blanc
- Service de pharmacie, hôpital Ambroise-Paré, 9, avenue Charles-de-Gaulle, 92104 Boulogne-Billancourt, France
| | - M-F De Maleissye
- Service de dermatologie, hôpital Ambroise-Paré, 9, avenue Charles-de-Gaulle, 92104 Boulogne-Billancourt, France
| | - S Bauler
- Service de pharmacie, hôpital Ambroise-Paré, 9, avenue Charles-de-Gaulle, 92104 Boulogne-Billancourt, France
| |
Collapse
|
246
|
Spagnolo F, Picasso V, Lambertini M, Ottaviano V, Dozin B, Queirolo P. Survival of patients with metastatic melanoma and brain metastases in the era of MAP-kinase inhibitors and immunologic checkpoint blockade antibodies: A systematic review. Cancer Treat Rev 2016; 45:38-45. [PMID: 26975020 DOI: 10.1016/j.ctrv.2016.03.003] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Revised: 03/01/2016] [Accepted: 03/02/2016] [Indexed: 12/22/2022]
Abstract
BACKGROUND The incidence of brain metastases (BM) in melanoma patients is common and associated with poor prognosis. MAP-kinase inhibitors and immunologic checkpoint blockade antibodies led to improved survival of metastatic melanoma patients; however, patients with BM are under-represented or excluded from the majority of clinical trials and the impact of new drugs on their survival is less clear. With the present systematic review, we aimed to analyze outcomes of patients with melanoma BM treated with the new drugs, both in the setting of phase I-II-III clinical trials and in the "real world". METHODS An electronic search was performed to identify studies reporting survival outcomes of patients with melanoma BM treated with MAP-kinase inhibitors and/or immunologic checkpoint blockade antibodies, regardless of study design. RESULTS Twenty-two studies were included for a total of 2153 patients. Median OS was 7.9 months in phase I-II-III trials and 7.7 months in "real world" studies. In clinical trials, median OS was 7.0 months for patients treated with immunotherapy and 7.9 months for patients treated with BRAF inhibitors. In "real world" studies, median OS was 4.3 months and 7.7 months for patients treated with immunotherapy and BRAF inhibitors, respectively. Evidence of clinical activity exists for both immunotherapy and MAP-kinase inhibitors. CONCLUSIONS MAP-kinase inhibitors and immunologic checkpoint blockade antibodies have clinical activity and may achieve improved OS in patients with metastatic melanoma and BM. These results support the inclusion of patients with BM in investigations of new agents and new treatment regimens for metastatic melanoma.
Collapse
Affiliation(s)
- Francesco Spagnolo
- Department of Plastic and Reconstructive Surgery, IRCCS AOU San Martino, IST Istituto Nazionale per la Ricerca sul Cancro, Genova, Italy.
| | - Virginia Picasso
- Department of Medical Oncology, U.O. Oncologia Medica 2, IRCCS AOU San Martino, IST Istituto Nazionale per la Ricerca sul Cancro, Genova, Italy
| | - Matteo Lambertini
- Department of Medical Oncology, U.O. Oncologia Medica 2, IRCCS AOU San Martino, IST Istituto Nazionale per la Ricerca sul Cancro, Genova, Italy
| | - Vincenzo Ottaviano
- Department of Plastic and Reconstructive Surgery, St George's Hospital, London, UK
| | - Beatrice Dozin
- Clinical Epidemiology Unit, IRCCS AOU San Martino, IST Istituto Nazionale per la Ricerca sul Cancro, Genova, Italy
| | - Paola Queirolo
- Department of Medical Oncology, U.O. Oncologia Medica 2, IRCCS AOU San Martino, IST Istituto Nazionale per la Ricerca sul Cancro, Genova, Italy
| |
Collapse
|
247
|
Ouellet D, Kassir N, Chiu J, Mouksassi MS, Leonowens C, Cox D, DeMarini DJ, Gardner O, Crist W, Patel K. Population pharmacokinetics and exposure-response of trametinib, a MEK inhibitor, in patients with BRAF V600 mutation-positive melanoma. Cancer Chemother Pharmacol 2016; 77:807-17. [PMID: 26940938 DOI: 10.1007/s00280-016-2993-y] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 02/15/2016] [Indexed: 11/30/2022]
Abstract
PURPOSE To characterize the pharmacokinetics of oral trametinib, a first in class MEK inhibitor, identify covariates, and describe the relationship between exposure and clinical effects in patients with BRAF V600 metastatic melanoma. EXPERIMENTAL DESIGN Trametinib concentrations obtained in three clinical studies were included in the population pharmacokinetic analysis. Trametinib 2 mg once daily was administered in the Phase 2 and 3 studies. The impact of exposure [trough (C min) or average concentration] on response rates and progression-free survival (PFS) was examined. RESULTS Plasma concentrations (n = 3120) obtained in 493 patients were described using a two-compartment model. Trametinib oral clearance was lower in women relative to men (1.26-fold) and increased with body weight. There was no significant effect of age, mild or moderate renal impairment, or mild hepatic impairment on oral clearance. Between-subject variability was low (24 %). The number of responders was consistent across median exposure range, although tended to be lower at trough concentration <10 ng/mL. Disease stage was found to be a significant predictor of response with a lower response rate in patients with disease stage of M1c. Lactate dehydrogenase was significant in the analysis of PFS. Patients with observed C min above the median had longer PFS than those below median based on Phase 2 study (median 10.6 ng/mL), while the effect of exposure was not statistically significant in the Phase 3 study (median 13.6 ng/mL). CONCLUSIONS No dosage adjustments are required with any of the covariates tested. Clinical efficacy was associated with trametinib trough concentrations greater than 10 ng/mL.
Collapse
Affiliation(s)
- Daniele Ouellet
- GlaxoSmithKline, Research Triangle Park, NC, USA. .,Janssen Research & Development, Spring House, PA, USA.
| | | | - Joannellyn Chiu
- GlaxoSmithKline, Research Triangle Park, NC, USA.,Inncelerex,, Jersey City, NJ, USA
| | | | - Cathrine Leonowens
- GlaxoSmithKline, Research Triangle Park, NC, USA.,Genentech, South San Francisco, CA, USA
| | - Donna Cox
- Incyte Corporation, Wilmington, DE, USA.,Teva Pharmaceuticals, Frazer, PA, USA
| | | | - Olivia Gardner
- GlaxoSmithKline, Research Triangle Park, NC, USA.,Janssen Research & Development, Spring House, PA, USA
| | - Wendy Crist
- Janssen Research & Development, Spring House, PA, USA.,Incyte Corporation, Wilmington, DE, USA
| | - Kiran Patel
- Janssen Research & Development, Spring House, PA, USA.,Incyte Corporation, Wilmington, DE, USA
| |
Collapse
|
248
|
Economic Evaluation of Ipilimumab in First Line Treatment of Advanced Melanoma in Italy. GLOBAL & REGIONAL HEALTH TECHNOLOGY ASSESSMENT 2016. [DOI: 10.5301/grhta.5000230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
249
|
Brummer GC, Bowen AR, Bowen GM. Merkel Cell Carcinoma: Current Issues Regarding Diagnosis, Management, and Emerging Treatment Strategies. Am J Clin Dermatol 2016; 17:49-62. [PMID: 26596990 DOI: 10.1007/s40257-015-0163-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Merkel cell carcinoma (MCC) is a rare but aggressive cutaneous tumor with a predilection for the head and neck of elderly Caucasian patients. Although much less common than melanoma, MCC has higher rates of sentinel lymph node involvement, local and regional recurrences, and mortality. The majority of MCC cases have been linked to the relatively newly discovered Merkel cell polyomavirus, which is a ubiquitous constituent of the skin flora. Recent discoveries regarding viral integration and carcinogenesis and the immunologic features of MCC have expanded the understanding of MCC. These discoveries have led to the development and application of emerging therapies such as somatostatin analogs, immune checkpoint inhibition, adoptive cell therapy, and other exciting possibilities for targeted therapy.
Collapse
|
250
|
Tang R, Xu X, Yang W, Yu W, Hou S, Xuan Y, Tang Z, Zhao S, Chen Y, Xiao X, Huang W, Guo W, Li M, Deng W. MED27 promotes melanoma growth by targeting AKT/MAPK and NF-κB/iNOS signaling pathways. Cancer Lett 2016; 373:77-87. [PMID: 26797421 DOI: 10.1016/j.canlet.2016.01.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 12/28/2015] [Accepted: 01/06/2016] [Indexed: 01/07/2023]
Abstract
The inhibitors of BRAF and MEK targeting MAPK signaling pathway provide a comparatively effective therapeutic strategy for melanoma caused by BRAF mutation. However, melanoma, especially metastatic melanoma, has become one of the most threatening malignancies. Thus, the identification of exact molecular mechanisms and the key components involved in such mechanisms is urgently needed in order to provide new therapeutic options for patients with melanoma. Here, we identified MED27 as a potential melanoma target and explored its role and the associated molecular mechanism involved in melanoma progression. MED27 was found to be highly expressed in melanoma cells and tumor tissues. Its silencing led to melanoma cell proliferation inhibition, cell cycle arrest and apoptosis induction accompanied by the inactivation of PI3K/AKT and MAPK/ERK signaling and the activation of Bax/Cyto-C/Caspase-dependent apoptotic pathway. In addition, silencing of MED27 led to the decrease of iNOS expression through inhibiting the activation of a serial of upstream key proteins of NF-κB signaling pathway and the translocation of p50/p65 from cytoplasm to nucleus. MED27 was also found to be able to interact with NF-κB and p300 and to be acetylated by p300. Furthermore, the results in a xenograft tumor model indicated that melanoma progression was effectively suppressed by MED27 knockdown accompanied by the down-regulation of p-AKT, p-ERK, p-MEK1/2, MMP-9, Bcl-2 and iNOS expressions in the tumor tissues. Taken together, our study not only demonstrated the new function of MED27 as an oncogenic protein and the associated molecular mechanisms involved in melanoma progression, but also provided a possibility for the development of MED27 as a new anticancer target in melanoma therapy.
Collapse
Affiliation(s)
- Ranran Tang
- Institute of Cancer Stem Cell & The Second Affiliated Hospital, Dalian Medical University, Dalian, China; Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Xiangdong Xu
- Department of Thyroid & Breast Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wenjing Yang
- Institute of Cancer Stem Cell & The Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Wendan Yu
- Institute of Cancer Stem Cell & The Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Shuai Hou
- Institute of Cancer Stem Cell & The Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Yang Xuan
- Institute of Cancer Stem Cell & The Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Zhipeng Tang
- Institute of Cancer Stem Cell & The Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Shilei Zhao
- Institute of Cancer Stem Cell & The Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Yiming Chen
- Institute of Cancer Stem Cell & The Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Xiangsheng Xiao
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Wenlin Huang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China; State Key Laboratory of Targeted Drug for Tumors of Guangdong Province, Guangzhou Double Bioproduct Inc., Guangzhou, China
| | - Wei Guo
- Institute of Cancer Stem Cell & The Second Affiliated Hospital, Dalian Medical University, Dalian, China.
| | - Man Li
- Institute of Cancer Stem Cell & The Second Affiliated Hospital, Dalian Medical University, Dalian, China.
| | - Wuguo Deng
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China; State Key Laboratory of Targeted Drug for Tumors of Guangdong Province, Guangzhou Double Bioproduct Inc., Guangzhou, China.
| |
Collapse
|