901
|
Montaudié H, Pradelli J, Passeron T, Lacour JP, Leroy S. Pulmonary sarcoid-like granulomatosis induced by nivolumab. Br J Dermatol 2016; 176:1060-1063. [PMID: 27291635 DOI: 10.1111/bjd.14808] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/18/2016] [Indexed: 01/04/2023]
Abstract
The use of antibodies against programmed death (PD)1, such as nivolumab and pembrolizumab, has dramatically improved the prognosis of patients with advanced melanoma. Nivolumab is also approved in advanced squamous cell nonsmall-cell lung cancer. These immunotherapies are associated with a unique set of toxicities termed immune-related adverse events, which are different from toxicities observed with conventional cytotoxic chemotherapy. We report the case of a 56-year-old man who was diagnosed with metastatic melanoma and who received nivolumab. One week after the second infusion, he developed pulmonary symptoms, dry eye syndrome and a bilateral swelling of the parotid glands. Investigations were negative for infection. The bronchoalveolar lavage differential cell count showed 32% lymphocytes with an increased CD4 : CD8 ratio, and bronchial biopsies revealed noncaseating epithelioid granulomas, without malignant cells. The clinical and radiological courses were rapidly favourable with oral corticosteroid. This case illustrates that sarcoidosis can be induced by nivolumab treatment. With the increasing use of anti-PD1 inhibitors in patients with advanced melanoma and squamous cell nonsmall-cell lung cancer, clinicians should be aware of this potential associated immune-related adverse event.
Collapse
Affiliation(s)
- H Montaudié
- Dermatology Department, Archet Hospital, Nice University Hospital, Nice, France
| | - J Pradelli
- Respiratory Department, Pasteur Hospital, Nice University Hospital, Nice, France
| | - T Passeron
- Dermatology Department, Archet Hospital, Nice University Hospital, Nice, France.,INSERM, U1065, Centre Méditerranéen de Médecine Moléculaire, Team 12, Nice, France
| | - J-P Lacour
- Dermatology Department, Archet Hospital, Nice University Hospital, Nice, France
| | - S Leroy
- Respiratory Department, Pasteur Hospital, Nice University Hospital, Nice, France
| |
Collapse
|
902
|
Rose AAN, Annis MG, Frederick DT, Biondini M, Dong Z, Kwong L, Chin L, Keler T, Hawthorne T, Watson IR, Flaherty KT, Siegel PM. MAPK Pathway Inhibitors Sensitize BRAF-Mutant Melanoma to an Antibody-Drug Conjugate Targeting GPNMB. Clin Cancer Res 2016; 22:6088-6098. [PMID: 27515299 PMCID: PMC6168941 DOI: 10.1158/1078-0432.ccr-16-1192] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 07/17/2016] [Accepted: 07/19/2016] [Indexed: 11/16/2022]
Abstract
PURPOSE To determine if BRAF and/or MEK inhibitor-induced GPNMB expression renders melanomas sensitive to CDX-011, an antibody-drug conjugate targeting GPNMB. EXPERIMENTAL DESIGN The Cancer Genome Atlas melanoma dataset was interrogated for a panel of MITF-regulated melanosomal differentiation antigens, including GPNMB. BRAF-mutant melanoma cell lines treated with BRAF or MEK inhibitors were assessed for GPNMB expression by RT-qPCR, immunoblot, and FACS analyses. Transient siRNA-mediated knockdown approaches were used to determine if MITF is requirement for treatment-induced GPNMB upregulation. GPNMB expression was analyzed in serial biopsies and serum samples from patients with melanoma taken before, during, and after disease progression on MAPK inhibitor treatment. Subcutaneous injections were performed to test the efficacy of MAPK inhibitors alone, CDX-011 alone, or their combination in suppressing melanoma growth. RESULTS A MITF-dependent melanosomal differentiation signature is associated with poor prognosis in patients with this disease. MITF is increased following BRAF and MEK inhibitor treatment and induces the expression of melanosomal differentiation genes, including GPNMB. GPNMB is expressed at the cell surface in MAPK inhibitor-treated melanoma cells and is also elevated in on-treatment versus pretreatment biopsies from melanoma patients receiving MAPK pathway inhibitors. Combining BRAF and/or MEK inhibitors with CDX-011, an antibody-drug conjugate targeting GPNMB, is effective in causing melanoma regression in preclinical animal models and delays the recurrent melanoma growth observed with MEK or BRAF/MEK inhibitor treatment alone. CONCLUSIONS The combination of MAPK pathway inhibitors with an antibody-drug conjugate targeting GPNMB is an effective therapeutic option for patients with melanoma. Clin Cancer Res; 22(24); 6088-98. ©2016 AACR.
Collapse
Affiliation(s)
- April A N Rose
- Goodman Cancer Research Centre, McGill University, Montréal, Québec, Canada
- Department of Medicine, McGill University, Montréal, Québec, Canada
| | - Matthew G Annis
- Goodman Cancer Research Centre, McGill University, Montréal, Québec, Canada
- Department of Medicine, McGill University, Montréal, Québec, Canada
| | | | - Marco Biondini
- Goodman Cancer Research Centre, McGill University, Montréal, Québec, Canada
- Department of Medicine, McGill University, Montréal, Québec, Canada
| | - Zhifeng Dong
- Goodman Cancer Research Centre, McGill University, Montréal, Québec, Canada
- Department of Medicine, McGill University, Montréal, Québec, Canada
| | - Lawrence Kwong
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Lynda Chin
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Institute for Applied Cancer Science, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | | | - Ian R Watson
- Goodman Cancer Research Centre, McGill University, Montréal, Québec, Canada
- Department of Biochemistry, McGill University, Montréal, Québec, Canada
| | - Keith T Flaherty
- Department of Surgical Oncology, Massachusetts General Hospital, Boston, Massachusetts
| | - Peter M Siegel
- Goodman Cancer Research Centre, McGill University, Montréal, Québec, Canada.
- Department of Medicine, McGill University, Montréal, Québec, Canada
- Department of Biochemistry, McGill University, Montréal, Québec, Canada
| |
Collapse
|
903
|
Rutkowski P. Introduction to the special issue of European Journal of Surgical Oncology: New roads in melanoma management. Eur J Surg Oncol 2016; 43:513-516. [PMID: 28034500 DOI: 10.1016/j.ejso.2016.12.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 12/06/2016] [Indexed: 12/27/2022] Open
Affiliation(s)
- P Rutkowski
- Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Department of Soft Tissue/Bone Sarcoma and Melanoma, Roentgena Str. 5, 02-781 Warsaw, Poland.
| |
Collapse
|
904
|
Chowdhary M, Patel KR, Danish HH, Lawson DH, Khan MK. BRAF inhibitors and radiotherapy for melanoma brain metastases: potential advantages and disadvantages of combination therapy. Onco Targets Ther 2016; 9:7149-7159. [PMID: 28003758 PMCID: PMC5161425 DOI: 10.2147/ott.s119428] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Melanoma is an aggressive malignancy that frequently spreads to the brain, resulting in rapid deterioration in both quality and quantity of life. Historically, treatment options for melanoma brain metastases (MBM) have predominantly consisted of surgery and radiotherapy. While these options can help provide local control, the majority of patients still develop intracranial progression. Indeed, novel therapeutic options, including molecularly targeted agents and immunotherapy, have improved outcomes and are now changing the role of radiotherapy. Up to 50% of melanomas contain an activating BRAF mutation, resulting in hyperactive cellular proliferation and survival. Drugs that target BRAF have been introduced for the treatment of metastatic melanoma and offer hope in improving disease outcomes; however, many of these trials either excluded or had a limited amount of patients with MBM. Recent studies have revealed that melanoma cell lines become more radiosensitive following BRAF inhibition, thus providing a potential synergistic mechanism when combining BRAF inhibitor (BRAFi) and radiotherapy. However, neurotoxicity concerns also exist with this combination. This article reviews the efficacy and limitations of BRAFi therapy for MBM, describes current evidence for combining BRAFis with radiation, discusses the rationale and evidence for combination modalities, and highlights emerging clinical trials specifically investigating this combination in MBM.
Collapse
Affiliation(s)
- Mudit Chowdhary
- Department of Radiation Oncology, Winship Cancer Institute, Emory University, Atlanta, GA
- Department of Radiation Oncology, Rush University Medical Center, Chicago, IL
| | - Kirtesh R Patel
- Department of Radiation Oncology, Winship Cancer Institute, Emory University, Atlanta, GA
| | - Hasan H Danish
- Department of Radiation Oncology, Winship Cancer Institute, Emory University, Atlanta, GA
| | - David H Lawson
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Mohammad K Khan
- Department of Radiation Oncology, Winship Cancer Institute, Emory University, Atlanta, GA
| |
Collapse
|
905
|
Evans J, Laking G, Strother M, Wang T, Metcalfe S, Blick G, Pauls R, Crausaz S. Mind the gap: An analysis of foregone health gains from unfunded cancer medicines in New Zealand. Semin Oncol 2016; 43:625-637. [DOI: 10.1053/j.seminoncol.2016.10.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 10/22/2016] [Indexed: 11/11/2022]
|
906
|
Atkinson V, Long GV, Menzies AM, McArthur G, Carlino MS, Millward M, Roberts-Thomson R, Brady B, Kefford R, Haydon A, Cebon J. Optimizing combination dabrafenib and trametinib therapy in BRAF mutation-positive advanced melanoma patients: Guidelines from Australian melanoma medical oncologists. Asia Pac J Clin Oncol 2016; 12 Suppl 7:5-12. [PMID: 27905182 DOI: 10.1111/ajco.12656] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
BRAF mutations occur commonly in metastatic melanomas and inhibition of mutant BRAF and the downstream kinase MEK results in rapid tumor regression and prolonged survival in patients. Combined therapy with BRAF and MEK inhibition improves response rate, progression free survival and overall survival compared with single agent BRAF inhibition, and reduces the skin toxicity that is seen with BRAF inhibitor monotherapy. However, this combination is associated with an increase in other toxicities, particularly drug-related pyrexia, which affects approximately 50% of patients treated with dabrafenib and trametinib (CombiDT). We provide guidance on managing adverse events likely to arise during treatment with combination BRAF and MEK inhibition with CombiDT: pyrexia, skin conditions, fatigue; and discuss management of CombiDT during surgery and radiotherapy. By improving tolerability and in particular preventing unnecessary treatment cessations or reduction in drug exposure, best outcomes can be achieved for patients undergoing CombiDT therapy.
Collapse
Affiliation(s)
- Victoria Atkinson
- Princess Alexandra Hospital, Greenslopes Private Hospital and University of Queensland, Brisbane, Queensland, Australia
| | - Georgina V Long
- Melanoma Institute Australia, Royal North Shore and Mater Hospitals, The University of Sydney, Sydney, New South Wales, Australia
| | - Alexander M Menzies
- Melanoma Institute Australia, Royal North Shore and Mater Hospitals, The University of Sydney, Sydney, New South Wales, Australia
| | - Grant McArthur
- Peter MacCallum Cancer Centre and Cabrini Health, Melbourne, Victoria, Australia
| | | | - Michael Millward
- School of Medicine and Pharmacology, University of Western Australia and Sir Charles Gairdner Hospital, Perth, Western Australia, Australia
| | | | - Benjamin Brady
- Peter MacCallum Cancer Centre and Cabrini Health, Melbourne, Victoria, Australia
| | - Richard Kefford
- Westmead Hospital and Macquarie University, Sydney, New South Wales, Australia
| | | | - Jonathan Cebon
- Olivia Newton John Cancer Wellness & Research Centre, Austin Health Melbourne, Victoria, Australia
| |
Collapse
|
907
|
Daste A, Chakiba C, Domblides C, Gross-goupil M, Quivy A, Ravaud A, Soubeyran P. Targeted therapy and elderly people: A review. Eur J Cancer 2016; 69:199-215. [DOI: 10.1016/j.ejca.2016.10.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Accepted: 10/05/2016] [Indexed: 11/26/2022]
|
908
|
Michels S, Wolf J. Stratified Treatment in Lung Cancer. Oncol Res Treat 2016; 39:760-766. [PMID: 27889787 DOI: 10.1159/000453406] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 11/15/2016] [Indexed: 11/19/2022]
Abstract
Even though great efforts have been made to improve chemotherapy-based treatment approaches for lung cancer, the prognosis of patients with advanced and metastasized disease remains particularly poor. In recent years, a growing number of genetic aberrations driving lung cancer have been identified. Targeted inhibition of some of these aberrations, most prominently mutated EGFR and ALK, by tyrosine kinase inhibitors has dramatically increased efficacy and tolerability of systemic lung cancer treatment in subsets of patients. However, the duration of response is limited due to the acquisition of molecular mechanisms of resistance to targeted treatment. Modern next-generation inhibitors aim to break resistance. A deep understanding of the mechanisms of treatment failure is imperative to the development of new approaches. In this review, we focus on the current status of stratified therapy in lung cancer and highlight new, potentially promising treatment approaches.
Collapse
Affiliation(s)
- Sebastian Michels
- Lung Cancer Group Cologne, Department I for Internal Medicine, Center for Integrated Oncology, University Hospital of Cologne, Cologne, Germany
| | | |
Collapse
|
909
|
Long GV, Grob JJ, Nathan P, Ribas A, Robert C, Schadendorf D, Lane SR, Mak C, Legenne P, Flaherty KT, Davies MA. Factors predictive of response, disease progression, and overall survival after dabrafenib and trametinib combination treatment: a pooled analysis of individual patient data from randomised trials. Lancet Oncol 2016; 17:1743-1754. [PMID: 27864013 DOI: 10.1016/s1470-2045(16)30578-2] [Citation(s) in RCA: 244] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 09/30/2016] [Accepted: 10/04/2016] [Indexed: 11/28/2022]
Abstract
BACKGROUND Dabrafenib plus trametinib treatment provides significant benefits over BRAF-inhibitor monotherapy in patients with BRAFV600E-mutant or BRAFV600K-mutant advanced melanoma; however, in many patients the disease progresses, leading to death. With many treatment options available, understanding clinical factors that predict long-term response and survival for treatments is important for optimisation of patient management. We aimed to identify clinical factors associated with long-term response and survival using pooled data from randomised trials of dabrafenib plus trametinib in patients with metastatic BRAF-mutant melanoma. METHODS We did a retrospective individual data analysis based on all published randomised trials that included treatment-naive patients with BRAFV600E-mutant or BRAFV600K-mutant metastatic melanoma who received the approved dose of dabrafenib 150 mg twice daily plus trametinib 2 mg once daily. Data were pooled from patients in the BRF113220 (part C; March 26, 2010, to Jan 15, 2015), COMBI-d (May 4, 2012, to Jan 12, 2015), and COMBI-v (June 4, 2012, to March 13, 2015) randomised trials. Patients with untreated brain metastases were not permitted to enrol in these trials. Baseline factors, identified a priori based on known melanoma clinical or prognostic characteristics, were analysed for association with progression-free survival and overall survival using univariate and multivariate analyses and assessed for hierarchical effect on outcomes using regression tree analyses. We also analysed factors identified after baseline, on treatment, and at progression, for associations with survival after progression. The trials included in this analysis are registered with ClinicalTrials.gov: BRF113220, number NCT01072175; COMBI-d, number NCT01584648; COMBI-v, number NCT01597908. FINDINGS 617 patients were included in this analysis with a median follow-up of 20·0 months (range 0-48·0, IQR 10·1-24·8); 396 patients had progression events (ie, disease progression or death) and 290 patients had died. Median progression-free survival (11·1 months [95% CI 9·7-12·9]), median overall survival (25·6 months [23·1-34·3]), 1-year progression-free survival (48% [44-52]) and overall survival (74% [71-78]), and 2-year progression-free survival (30% [26-34]) and overall survival (53% [49-57]) were consistent with those in the individual trials. Patients with normal lactate dehydrogenase (LDH) concentration and fewer than three organ sites containing metastases (n=237) had the longest 1-year progression-free survival (68% [95% CI 62-74]) and overall survival (90% [87-94]) and 2-year progression-free survival (46% [40-54]) and overall survival (75% [70-81]), whereas patients with LDH concentration at least two times the upper limit of normal (n=70) had the shortest 1-year progression-free survival (8% [3-19]) and overall survival (40% [29-55]) and 2-year progression-free survival (2% [0-13]) and overall survival (7% [3-19]). Of patients with disease progression (n=379), survival after progression was longest in those with progression in baseline or new non-CNS lesions (n=205; median 10·0 months [95% CI 7·9-12·0]) and shortest in those with new CNS lesions or concurrent progression in baseline and new lesions (n=171; median 4·0 months [3·5-4·9]). INTERPRETATION Several patient and clinical characteristics at and after baseline are associated with outcomes with dabrafenib plus trametinib, and durable benefit is possible with targeted treatment in defined patient subsets. FUNDING Novartis.
Collapse
Affiliation(s)
- Georgina V Long
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia; Royal North Shore and Mater Hospitals, Sydney, Australia.
| | - Jean-Jacques Grob
- Service de Dermatologie, Centre Hospitalo-Universitaire Timone, Aix Marseille Université, Marseille CEDEX 05, France
| | | | - Antoni Ribas
- Department of Medicine, Hematology/Oncology, UCLA Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Caroline Robert
- Gustave Roussy, Département de Médecine Oncologique, Service de Dermatologie et Université Paris-Sud, Faculté de Médecine, Villejuif, France
| | - Dirk Schadendorf
- Department of Dermatology, University Hospital Essen, Essen, Germany; German Cancer Consortium, Heidelberg, Germany
| | - Stephen R Lane
- Biostatistics, Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA
| | - Carmen Mak
- Biostatistics, Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA
| | | | - Keith T Flaherty
- Developmental Therapeutics and Melanoma Programs, Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | - Michael A Davies
- Melanoma Medical Oncology and Systems Biology, The University of Texas MDAnderson Cancer Center, Houston, TX, USA
| |
Collapse
|
910
|
Combinatorial immunotherapy for melanoma. Cancer Gene Ther 2016; 24:141-147. [PMID: 27834353 DOI: 10.1038/cgt.2016.56] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 09/29/2016] [Indexed: 12/18/2022]
Abstract
Melanoma has been a long-standing focal point for immunotherapy development. In this review, we explore the evolution of melanoma treatments with particular attention to the history and recent advances in melanoma immunotherapy. We also discuss novel combinations of these modalities and their potential to offer novel therapeutic options for patients with advanced melanoma.
Collapse
|
911
|
Khalil DN, Postow MA, Ibrahim N, Ludwig DL, Cosaert J, Kambhampati SRP, Tang S, Grebennik D, Kauh JSW, Lenz HJ, Flaherty KT, Hodi FS, Lawrence DP, Wolchok JD. An Open-Label, Dose-Escalation Phase I Study of Anti-TYRP1 Monoclonal Antibody IMC-20D7S for Patients with Relapsed or Refractory Melanoma. Clin Cancer Res 2016; 22:5204-5210. [PMID: 27797971 PMCID: PMC5117650 DOI: 10.1158/1078-0432.ccr-16-1241] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Accepted: 08/04/2016] [Indexed: 11/16/2022]
Abstract
PURPOSE Tyrosinase-related protein-1 (TYRP1) is a transmembrane glycoprotein that is specifically expressed in melanocytes and melanoma cells. Preclinical data suggest that mAbs targeting TYRP1 confer antimelanoma activity. IMC-20D7S is a recombinant human IgG1 mAb targeting TYRP1. Here, we report the first-in-human phase I/Ib trial of IMC-20D7S. EXPERIMENTAL DESIGN The primary objective of this study was to establish the safety profile and the MTD of IMC-20D7S. Patients with advanced melanoma who progressed after or during at least one line of treatment or for whom standard therapy was not indicated enrolled in this standard 3 + 3 dose-escalation, open-label study. IMC-20D7S was administered intravenously every 2 or 3 weeks. RESULTS Twenty-seven patients were enrolled. The most common adverse events were fatigue and constipation experienced by nine (33%) and eight (30%) patients, respectively. There were no serious adverse events related to treatment, no discontinuations of treatment due to adverse events, and no treatment-related deaths. Given the absence of dose-limiting toxicities, an MTD was not defined, but a provisional MTD was established at the 20 mg/kg every 2-week dose based on serum concentration and safety data. One patient experienced a complete response. A disease control rate, defined as stable disease or better, of 41% was observed. CONCLUSION IMC-20D7S is well tolerated among patients with advanced melanoma with evidence of antitumor activity. Further investigation of this agent as monotherapy in selected patients or as part of combination regimens is warranted. Clin Cancer Res; 22(21); 5204-10. ©2016 AACR.
Collapse
Affiliation(s)
- Danny N Khalil
- Memorial Sloan Kettering Cancer Center, Ludwig Center for Cancer Immunotherapy, New York, New York
| | - Michael A Postow
- Memorial Sloan Kettering Cancer Center, Ludwig Center for Cancer Immunotherapy, New York, New York
| | | | | | | | | | | | | | | | - Heinz-Josef Lenz
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California
| | | | - F Stephen Hodi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | | | - Jedd D Wolchok
- Memorial Sloan Kettering Cancer Center, Ludwig Center for Cancer Immunotherapy, New York, New York.
| |
Collapse
|
912
|
Jamieson D, Griffin MJ, Sludden J, Drew Y, Cresti N, Swales K, Merriman M, Allen R, Bevan P, Buerkle M, Mala C, Coyle V, Rodgers L, Dean E, Greystoke A, Banerji U, Wilson RH, Evans TRJ, Anthoney A, Ranson M, Boddy AV, Plummer R. A phase I pharmacokinetic and pharmacodynamic study of the oral mitogen-activated protein kinase kinase (MEK) inhibitor, WX-554, in patients with advanced solid tumours. Eur J Cancer 2016; 68:1-10. [PMID: 27693888 DOI: 10.1016/j.ejca.2016.08.026] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Revised: 08/15/2016] [Accepted: 08/27/2016] [Indexed: 11/15/2022]
Abstract
PURPOSE We performed a multi-centre phase I study to assess the safety, pharmacokinetics (PK) and pharmacodynamics (PD) of the orally available small molecule mitogen-activated protein kinase kinase (MEK) 1/2 inhibitor, WX-554, and to determine the optimal biological dose for subsequent trials. EXPERIMENTAL DESIGN Patients with treatment-refractory, advanced solid tumours, with adequate performance status and organ function were recruited to a dose-escalation study in a standard 3 + 3 design. The starting dose was 25 mg orally once weekly with toxicity, PK and PD guided dose-escalation with potential to explore alternative schedules. RESULTS Forty-one patients with advanced solid tumours refractory to standard therapies and with adequate organ function were recruited in eight cohorts up to doses of 150 mg once weekly and 75 mg twice weekly. No dose-limiting toxicities were observed during the study, and a maximum tolerated dose (MTD) was not established. The highest dose cohorts demonstrated sustained inhibition of extracellular signal-regulated kinase (ERK) phosphorylation in peripheral blood mononuclear cells following ex-vivo phorbol 12-myristate 13-acetate stimulation. There was a decrease of 70 ± 26% in mean phosphorylated (p)ERK in C1 day 8 tumour biopsies when compared with pre-treatment tumour levels in the 75 mg twice a week cohort. Prolonged stable disease (>6 months) was seen in two patients, one with cervical cancer and one with ampullary carcinoma. CONCLUSIONS WX-554 was well tolerated, and an optimal biological dose was established for further investigation in either a once or twice weekly regimens. The recommended phase 2 dose is 75 mg twice weekly.
Collapse
Affiliation(s)
- David Jamieson
- Northern Institute for Cancer Research, Paul O'Gorman Building, Medical School, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Melanie J Griffin
- Northern Institute for Cancer Research, Paul O'Gorman Building, Medical School, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Julieann Sludden
- Northern Institute for Cancer Research, Paul O'Gorman Building, Medical School, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Yvette Drew
- Northern Institute for Cancer Research, Paul O'Gorman Building, Medical School, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK; Northern Centre for Cancer Care, Newcastle Freeman Hospital, Newcastle upon Tyne, NE7 7DN, UK
| | - Nicola Cresti
- Northern Centre for Cancer Care, Newcastle Freeman Hospital, Newcastle upon Tyne, NE7 7DN, UK
| | - Karen Swales
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, UK
| | | | | | - Paul Bevan
- WILEX AG, Grillparzerstr. 18, 81675, Munich, Germany
| | | | - Carola Mala
- WILEX AG, Grillparzerstr. 18, 81675, Munich, Germany
| | - Vicky Coyle
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK; Northern Ireland Cancer Center, Belfast City Hospital, Belfast, UK
| | - Lisa Rodgers
- Beatson West of Scotland Cancer Centre, University of Glasgow, Glasgow, G12 OYN, UK
| | - Emma Dean
- The Christie NHS Foundation Trust, The University of Manchester, Manchester, M20 4BX, UK
| | - Alastair Greystoke
- Northern Institute for Cancer Research, Paul O'Gorman Building, Medical School, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK; Northern Centre for Cancer Care, Newcastle Freeman Hospital, Newcastle upon Tyne, NE7 7DN, UK
| | - Udai Banerji
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, UK
| | - Richard H Wilson
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK; Northern Ireland Cancer Center, Belfast City Hospital, Belfast, UK
| | - T R Jeffery Evans
- Beatson West of Scotland Cancer Centre, University of Glasgow, Glasgow, G12 OYN, UK
| | - Alan Anthoney
- St. James's Institute of Oncology, Beckett Street, Leeds, LS9 7TF, UK
| | - Malcolm Ranson
- The Christie NHS Foundation Trust, The University of Manchester, Manchester, M20 4BX, UK
| | - Alan V Boddy
- Northern Institute for Cancer Research, Paul O'Gorman Building, Medical School, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Ruth Plummer
- Northern Institute for Cancer Research, Paul O'Gorman Building, Medical School, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK; Northern Centre for Cancer Care, Newcastle Freeman Hospital, Newcastle upon Tyne, NE7 7DN, UK.
| |
Collapse
|
913
|
Brown NF, Carter T, Mulholland P. Dabrafenib in BRAFV600-mutated anaplastic pleomorphic xanthoastrocytoma. CNS Oncol 2016; 6:5-9. [PMID: 27781490 DOI: 10.2217/cns-2016-0031] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Pleomorphic xanthoastrocytoma (PXA) is a rare brain tumor. Anaplastic features are found in 20-30% of cases of PXA and are associated with poor outcomes. Typical treatment is with gross total resection, followed by radiation therapy and cytotoxic chemotherapy at relapse. BRAFV600 mutations have been identified in 38-60% of patients with PXA. Several case reports and small case series have identified clinical benefit with BRAF inhibition in patients with BRAFV600-mutated PXA. We report the second published case of successful treatment with the BRAF inhibitor dabrafenib in a female patient with relapsed anaplastic PXA with a BRAFV600 mutation, and the first published case of dabrafinib treatment following intolerance to vemurafenib.
Collapse
Affiliation(s)
- Nicholas F Brown
- Department of Oncology, University College London Hospitals, 235 Euston Road, London NW1 2BU, UK.,UCL Cancer Institute, University College London, 72 Huntley Street, London WC1E 6DD, UK
| | - Thomas Carter
- UCL Cancer Institute, University College London, 72 Huntley Street, London WC1E 6DD, UK
| | - Paul Mulholland
- Department of Oncology, University College London Hospitals, 235 Euston Road, London NW1 2BU, UK.,UCL Cancer Institute, University College London, 72 Huntley Street, London WC1E 6DD, UK
| |
Collapse
|
914
|
Cohen JV, Tawbi H, Margolin KA, Amravadi R, Bosenberg M, Brastianos PK, Chiang VL, de Groot J, Glitza IC, Herlyn M, Holmen SL, Jilaveanu LB, Lassman A, Moschos S, Postow MA, Thomas R, Tsiouris JA, Wen P, White RM, Turnham T, Davies MA, Kluger HM. Melanoma central nervous system metastases: current approaches, challenges, and opportunities. Pigment Cell Melanoma Res 2016; 29:627-642. [PMID: 27615400 DOI: 10.1111/pcmr.12538] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 09/06/2016] [Indexed: 12/17/2022]
Abstract
Melanoma central nervous system metastases are increasing, and the challenges presented by this patient population remain complex. In December 2015, the Melanoma Research Foundation and the Wistar Institute hosted the First Summit on Melanoma Central Nervous System (CNS) Metastases in Philadelphia, Pennsylvania. Here, we provide a review of the current status of the field of melanoma brain metastasis research; identify key challenges and opportunities for improving the outcomes in patients with melanoma brain metastases; and set a framework to optimize future research in this critical area.
Collapse
Affiliation(s)
- Justine V Cohen
- Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA
| | - Hussain Tawbi
- Department of Melanoma, Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kim A Margolin
- Department of Medical Oncology & Therapeutics Research, City of Hope Cancer Center, Duarte, CA, USA
| | - Ravi Amravadi
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | | | | | | | - John de Groot
- Division of Neuro-Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - Isabella C Glitza
- Department of Melanoma, Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Meenhard Herlyn
- Melanoma Research Center, The Wistar Institute, Philadelphia, PA, USA
| | - Sheri L Holmen
- Huntsman Cancer Institute, University of Utah Health Sciences Center, Salt Lake City, UT, USA
| | | | - Andrew Lassman
- Department of Neurology & Herbert Irving Comprehensive, Cancer Center, Columbia University Medical Center, New York, NY, USA
| | - Stergios Moschos
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Michael A Postow
- Department of Oncology, Memorial Sloan Kettering Cancer Center and Weill Cornell Medicine, New York, NY, USA
| | - Reena Thomas
- Division of Neuro-Oncology, Department of Neurology, Stanford University, Stanford, CA, USA
| | - John A Tsiouris
- Department of Radiology, New York-Presbyterian Hospital - Weill Cornell Medicine, New York, NY, USA
| | - Patrick Wen
- Department of Neuro-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Richard M White
- Department of Cancer Biology & Genetics, Memorial Sloan Kettering Cancer Center and Weill Cornell Medicine, New York, NY, USA
| | | | - Michael A Davies
- Department of Melanoma, Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | |
Collapse
|
915
|
Cohen R, Svrcek M, Dreyer C, Cervera P, Duval A, Pocard M, Fléjou JF, de Gramont A, André T. New Therapeutic Opportunities Based on DNA Mismatch Repair and BRAF Status in Metastatic Colorectal Cancer. Curr Oncol Rep 2016; 18:18. [PMID: 26861657 DOI: 10.1007/s11912-016-0504-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Recently, colorectal cancer (CRC) subtyping consortium identified four consensus molecular subtypes (CMS1-4). CMS1 is enriched for deficient mismatch repair (dMMR) and BRAF (V600E) tumors. Intriguingly, this subtype has better relapse-free survival but worse overall survival after relapse compared with the other subtypes. Growing evidence is accumulating on the benefit of specific therapeutic strategies such as immune checkpoint inhibition therapy in dMMR tumors and mitogen-activated protein kinase (MAPK) pathway targeted therapy in tumors harboring BRAF (V600E) mutation. After reviewing dMMR prognostic value, immune checkpoints as major targets for dMMR carcinomas will be highlighted. Following, BRAF (V600E) prognostic impact will be reviewed and therapeutic strategies with the combination of cytotoxic agents and especially the combinations of BRAF and MAPK inhibitors will be discussed.
Collapse
Affiliation(s)
- Romain Cohen
- Department of Medical Oncology, Hospital Saint-Antoine, Assistance publique-Hôpitaux de Paris, 184, rue du Faubourg-Saint-Antoine, 75012, Paris, France
| | - Magali Svrcek
- Department of Pathology, Hospital Saint-Antoine, APHP, 184 rue du Faubourg Saint-Antoine, Paris, 75012, France.,University Pierre et Marie Curie (UMPC), Paris VI, 4 Place Jussieu, Paris, 75005, France
| | - Chantal Dreyer
- Department of Medical Oncology, Hospital Saint-Antoine, Assistance publique-Hôpitaux de Paris, 184, rue du Faubourg-Saint-Antoine, 75012, Paris, France
| | - Pascale Cervera
- Department of Pathology, Hospital Saint-Antoine, APHP, 184 rue du Faubourg Saint-Antoine, Paris, 75012, France.,University Pierre et Marie Curie (UMPC), Paris VI, 4 Place Jussieu, Paris, 75005, France
| | - Alex Duval
- INSERM, Unité Mixte de Recherche Scientifique 938, Centre de Recherche Saint-Antoine, Equipe "Instabilité des Microsatellites et Cancers," Equipe labellisée par la Ligue Nationale contre le Cancer, 184 rue du Faubourg Saint-Antoine, Paris, 75012, France
| | - Marc Pocard
- GERCOR, Oncology Multidisciplinary Group, 151 rue du Faubourg Saint Antoine, Paris, 75011, France.,Departement of Digestive and Oncologic Surgery, Hospital Lariboisière, APHP, 2 rue Ambroise Paré, Paris, 75010, France
| | - Jean-François Fléjou
- Department of Pathology, Hospital Saint-Antoine, APHP, 184 rue du Faubourg Saint-Antoine, Paris, 75012, France.,University Pierre et Marie Curie (UMPC), Paris VI, 4 Place Jussieu, Paris, 75005, France
| | - Aimery de Gramont
- GERCOR, Oncology Multidisciplinary Group, 151 rue du Faubourg Saint Antoine, Paris, 75011, France.,Department of Medical Oncology, Institut Hospitalier Franco-Britannique, 4 rue Kléber, 92300, Levallois-Perret, France
| | - Thierry André
- Department of Medical Oncology, Hospital Saint-Antoine, Assistance publique-Hôpitaux de Paris, 184, rue du Faubourg-Saint-Antoine, 75012, Paris, France. .,University Pierre et Marie Curie (UMPC), Paris VI, 4 Place Jussieu, Paris, 75005, France. .,INSERM, Unité Mixte de Recherche Scientifique 938, Centre de Recherche Saint-Antoine, Equipe "Instabilité des Microsatellites et Cancers," Equipe labellisée par la Ligue Nationale contre le Cancer, 184 rue du Faubourg Saint-Antoine, Paris, 75012, France. .,GERCOR, Oncology Multidisciplinary Group, 151 rue du Faubourg Saint Antoine, Paris, 75011, France.
| |
Collapse
|
916
|
Dagogo-Jack I, Gill CM, Cahill DP, Santagata S, Brastianos PK. Treatment of brain metastases in the modern genomic era. Pharmacol Ther 2016; 170:64-72. [PMID: 27773784 DOI: 10.1016/j.pharmthera.2016.10.011] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Development of brain metastasis (BM) portends a dismal prognosis for patients with cancer. Melanomas and carcinomas of the lung, breast, and kidney are the most common malignancies to metastasize to the brain. Recent advances in molecular genetics have enabled the identification of actionable, clinically relevant genetic alterations within primary tumors and their corresponding metastases. Adoption of genotype-guided treatment strategies for the management of systemic malignancy has resulted in dramatic and durable responses. Unfortunately, despite these therapeutic advances, central nervous system (CNS) relapses are not uncommon. Although these relapses have historically been attributed to limited blood brain barrier penetration of anti-neoplastic agents, recent work has demonstrated genetic heterogeneity such that metastatic sites, including BM, harbor relevant genetic alterations that are not present in primary tumor biopsies. This improved insight into molecular mechanisms underlying site specific recurrences can inform strategies for targeting these oncogenic drivers. Thus, development of rational, genomically guided CNS-penetrant therapies is crucial for ongoing therapeutic success.
Collapse
Affiliation(s)
- Ibiayi Dagogo-Jack
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States; Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Corey M Gill
- Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States; Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Daniel P Cahill
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Sandro Santagata
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Priscilla K Brastianos
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States; Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States; Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
917
|
Liang R, Meiser B, Smith S, Kasparian N, Lewis C, Chin M, Long G, Ward R, Menzies A, Harris-Wai J, Kaur R. Advanced cancer patients’ attitudes towards, and experiences with, screening for somatic mutations in tumours: a qualitative study. Eur J Cancer Care (Engl) 2016; 26. [DOI: 10.1111/ecc.12600] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/13/2016] [Indexed: 12/14/2022]
Affiliation(s)
- R. Liang
- UNSW Medicine; University of New South Wales; Sydney NSW Australia
| | - B. Meiser
- Psychosocial Research Group; Prince of Wales Clinical School; Sydney NSW Australia
| | - S. Smith
- Psychosocial Research Group; Prince of Wales Clinical School; Sydney NSW Australia
| | - N.A. Kasparian
- Discipline of Paediatrics; School of Women's and Children's Health; UNSW Medicine; University of New South Wales; Sydney NSW Australia
| | - C.R. Lewis
- Department of Medical Oncology; Prince of Wales Hospital; Randwick NSW Australia
- Prince of Wales Clinical School; Randwick NSW Australia
| | - M. Chin
- Department of Medical Oncology; Prince of Wales Hospital; Randwick NSW Australia
- Prince of Wales Clinical School; Randwick NSW Australia
| | - G.V. Long
- Melanoma Institute of Australia; The University of Sydney; Mater and Royal North Shore Hospital; Sydney NSW Australia
| | - R. Ward
- Prince of Wales Clinical School; Randwick NSW Australia
- University of Queensland; Brisbane QLD Australia
| | - A.M. Menzies
- Melanoma Institute of Australia; The University of Sydney; Mater and Royal North Shore Hospital; Sydney NSW Australia
| | - J.N. Harris-Wai
- Division of Research; Kaiser Permanente Northern California; Oakland CA USA
| | - R. Kaur
- Psychosocial Research Group; Prince of Wales Clinical School; Sydney NSW Australia
| |
Collapse
|
918
|
Palumbo G, Di Lorenzo G, Ottaviano M, Damiano V. The future of melanoma therapy: developing new drugs and improving the use of old ones. Future Oncol 2016; 12:2531-2534. [PMID: 27715206 DOI: 10.2217/fon-2015-0045] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Giuliano Palumbo
- Medical Oncology Unit, Department of Clinical Medicine, Federico II University, 80138 Naples, Italy
| | - Giuseppe Di Lorenzo
- Medical Oncology Unit, Department of Clinical Medicine, Federico II University, 80138 Naples, Italy
| | - Margaret Ottaviano
- Medical Oncology Unit, Department of Clinical Medicine, Federico II University, 80138 Naples, Italy
| | - Vincenzo Damiano
- Medical Oncology Unit, Department of Clinical Medicine, Federico II University, 80138 Naples, Italy
| |
Collapse
|
919
|
Abstract
OBJECTIVE To review and summarize data on cobimetinib, which was approved by the US Food and Drug Administration (FDA) in November 2015 for use in combination with vemurafenib for unresectable or metastatic melanoma with a BRAFV600E or V600K mutation. DATA SOURCES A literature search using PubMed was conducted using the terms cobimetinib, MEK inhibitor, and melanoma from January 2000 to June 2016. STUDY SELECTION AND DATA EXTRACTION The literature search was confined to human studies published in English. Trials of cobimetinib for melanoma were prioritized. DATA SYNTHESIS Cobimetinib is a reversible inhibitor of MEK1 and MEK2. Its FDA approval was based on a phase III, randomized trial of vemurafenib monotherapy (n = 248) or vemurafenib and cobimetinib (n = 247) in unresectable stage IIIC or IV melanoma with a BRAFV600 mutation. Cobimetinib was administered as 60 mg orally daily for 21 days/7 days off, whereas vemurafenib was administered as 960 mg twice daily. Vemurafenib and cobimetinib were associated with an objective response rate of 68%, and median progression-free survival of 9.9 months. The overall survival was not reached at the time of first interim analysis. Clinically relevant grade ≥3 adverse events were diarrhea (6%), rash (6%), photosensitivity (2%), elevated liver function tests (LFTs) (8%-12%), increased creatine kinase (11%), and retinal detachment (3%). CONCLUSION Cobimetinib combined with vemurafenib is an alternative BRAF/MEK inhibitor therapy for unresectable or metastatic melanoma with BRAFV600 mutation. The role of cobimetinib in melanoma and other solid tumors is likely to expand as the results from ongoing studies become available.
Collapse
|
920
|
de Langen AJ, Smit EF. Therapeutic approach to treating patients with BRAF-mutant lung cancer: latest evidence and clinical implications. Ther Adv Med Oncol 2016; 9:46-58. [PMID: 28203297 DOI: 10.1177/1758834016670555] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Lung adenocarcinoma is known for its high rate of somatic mutations and genomic rearrangements. The identification of epidermal growth factor receptor (EGFR) mutations and anaplastic lymphoma kinase (ALK) rearrangements that sensitize tumors to specific drugs has changed the therapeutic approach and prognosis in these molecularly-defined subgroups. Several other key genetic alterations have been identified, of which BRAF mutations are found in 4% of non-small cell lung cancer (NSCLC) cases. Targeted drugs against BRAF and downstream MEK were recently approved for the treatment of BRAF-positive melanoma and have entered clinical evaluation in NSCLC. In this review we discuss the latest evidence on the treatment of BRAF-mutated NSCLC, including tumor biology, targeted treatment with BRAF and MEK inhibitors, therapeutic resistance and strategies to overcome resistance.
Collapse
Affiliation(s)
- Adrianus J de Langen
- Department of Pulmonary Diseases, VU University Medical Center, De Boelelaan 1117, 1007 MB Amsterdam, The Netherlands
| | - Egbert F Smit
- Department of Pulmonary Diseases, VU University Medical Center, and Department of Thoracic Oncology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| |
Collapse
|
921
|
Kruse V, Boterberg T, Acou M, Creytens D, Van den Broecke C, Brochez L, Hallaert G. Successful strategy to treat a solitary cystic melanoma brain metastasis. J Eur Acad Dermatol Venereol 2016; 31:e216-e217. [PMID: 27684056 DOI: 10.1111/jdv.13981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Affiliation(s)
- V Kruse
- Department of Medical Oncology, Ghent University Hospital, Gent, Belgium
| | - T Boterberg
- Department of Radiation Oncology, Ghent University Hospital, Gent, Belgium
| | - M Acou
- Department of Neuroradiology, Ghent University Hospital, Gent, Belgium
| | - D Creytens
- Department of Pathology, Ghent University Hospital, Gent, Belgium
| | | | - L Brochez
- Department of Dermatology, Ghent University Hospital, Gent, Belgium
| | - G Hallaert
- Department of Neurosurgery, Ghent University Hospital, Gent, Belgium
| |
Collapse
|
922
|
Amdahl J, Chen L, Delea TE. Network Meta-analysis of Progression-Free Survival and Overall Survival in First-Line Treatment of BRAF Mutation-Positive Metastatic Melanoma. Oncol Ther 2016; 4:239-256. [PMID: 28261653 PMCID: PMC5315084 DOI: 10.1007/s40487-016-0030-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Indexed: 01/23/2023] Open
Abstract
INTRODUCTION The present study aimed to inform an economic evaluation of dabrafenib and trametinib combination as first-line treatment of metastatic melanoma in a Canadian setting. A network meta-analysis was conducted to estimate hazard ratios (HRs) for progression-free survival (PFS)and overall survival (OS) of dabrafenib plus trametinib versus other first-line treatments of BRAF mutation-positive metastatic melanoma including dabrafenib, trametinib, vemurafenib, ipilimumab, and dacarbazine (DTIC). METHODS HRs for PFS and OS were from randomized controlled trials identified from systematic literature reviews. HRs for PFS and OS (adjusted for crossover as appropriate) were analyzed using multivariate and univariate Bayesian network meta-analysis. RESULTS In multivariate network-meta analyses (HRs for PFS and OS estimated simultaneously to account for the correlation of treatment effects on PFS and OS), HRs (95% credible interval) for PFS and OS favored dabrafenib plus trametinib [PFS: 0.23 (0.18-0.29) versus DTIC, 0.32 (0.24-0.42) versus ipilimumab plus DTIC, 0.52 (0.32-0.83) versus trametinib, 0.57 (0.48-0.69) versus vemurafenib, and 0.59 (0.50-0.71) versus dabrafenib]; OS [0.41 (0.29-0.56) versus DTIC, 0.52 (0.38-0.71) versus ipilimumab plus DTIC, 0.68 (0.47-0.95) versus trametinib, 0.69 (0.57-0.84) versus vemurafenib, and 0.72 (0.60-0.85) versus dabrafenib]. The beneficial effects on OS of dabrafenib plus trametinib versus ipilimumab plus DTIC and versus trametinib were attenuated when HRs were estimated using univariate network meta-analysis (HRs for PFS and OS estimated separately). CONCLUSION This analysis demonstrates improved PFS and OS with dabrafenib + trametinib versus dabrafenib, trametinib, vemurafenib, ipilimumab plus DTIC, and DTIC as first-line treatment for patients with BRAF mutation-positive metastatic melanoma. FUNDING Novartis Pharmaceuticals.
Collapse
Affiliation(s)
- Jordan Amdahl
- Policy Analysis Inc. (PAI), 4 Davis Court, Brookline, MA 02445 USA
| | - Lei Chen
- Novartis Pharmaceuticals, East Hanover, NJ USA
| | - Thomas E Delea
- Policy Analysis Inc. (PAI), 4 Davis Court, Brookline, MA 02445 USA
| |
Collapse
|
923
|
Luu C, Khushalani NI, Zager JS. Intralesional and systemic immunotherapy for metastatic melanoma. Expert Opin Biol Ther 2016; 16:1491-1499. [DOI: 10.1080/14712598.2016.1233961] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
924
|
Rauschenberg R, Garzarolli M, Dietrich U, Beissert S, Meier F. Systemic therapy of metastatic melanoma. J Dtsch Dermatol Ges 2016; 13:1223-35; quiz 1236-7. [PMID: 26612791 DOI: 10.1111/ddg.12891] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
For patients with metastatic melanoma, there are currently several effective therapeutic options. The BRAF inhibitors vemurafenib and dabrafenib are characterized by rapid tumor control and high response rates. In combination with one of the two MEK inhibitors trametinib and cobimetinib, they achieve response rates (CR + PR, complete plus partial remissions) of 70%, while delaying the development of treatment resistance, as well as a median overall survival of > 2 years with tolerable side effects. Showing long-term survival rates of approximately 20%, the anti-CTLA-4 antibody ipilimumab is the first substance that has led to a significant prolongation of overall survival in patients with metastatic melanoma. However, delayed treatment response and severe immune-mediated side effects may pose limitations to its therapeutic benefit. Usually well tolerated, anti-PD-1 antibody monotherapy using nivolumab and pembrolizumab has yielded response rates (CR + PR) of up to 45% and one-year survival rates of > 70%. The combination of ipilimumab and nivolumab has shown response rates of up to 58% and a median progression-free survival of > 11 months. While this combination is expected to result in a rapid and long-lasting response, this potential benefit comes at the expense of a high level of toxicity. Strategies for treatment sequencing and treatment combinations are currently being investigated in clinical studies. Overall, the prognosis for patients with metastatic melanoma has significantly improved. With long-term survival a possibility, not only acute but also long-term therapeutic side effects must be taken into account.
Collapse
Affiliation(s)
- Ricarda Rauschenberg
- Department of Dermatology, Carl Gustav Carus University Hospital at the Technical University Dresden, Dresden, Germany
| | - Marlene Garzarolli
- Department of Dermatology, Carl Gustav Carus University Hospital at the Technical University Dresden, Dresden, Germany
| | - Ursula Dietrich
- Department of Dermatology, Carl Gustav Carus University Hospital at the Technical University Dresden, Dresden, Germany
| | - Stefan Beissert
- Department of Dermatology, Carl Gustav Carus University Hospital at the Technical University Dresden, Dresden, Germany
| | - Friedegund Meier
- Department of Dermatology, Carl Gustav Carus University Hospital at the Technical University Dresden, Dresden, Germany
| |
Collapse
|
925
|
Abstract
BACKGROUND Several subsets of non-small-cell lung cancer (NSCLC) are defined by molecular alterations acting as tumor drivers, some of them being currently therapeutically actionable. The rat sarcoma (RAS)-rapidly accelerated fibrosarcoma (RAF)-mitogen-activated protein/extracellular signal-regulated kinase kinase (MEK)-extracellular signal-regulated kinase (ERK) pathway constitutes an attractive potential target, as v-Raf murine sarcoma viral oncogene homolog B (BRAF) mutations occur in 2-4% of NSCLC adenocarcinoma. METHODS Here, we review the latest clinical data on BRAF serine/threonine kinase inhibitors in NSCLC. RESULTS Treatment of V600E BRAF-mutated NSCLC with BRAF inhibitor monotherapy demonstrated encouraging antitumor activity. Combination of BRAF and MEK inhibitors using dabrafenib and trametinib is under evaluation. Preliminary data suggest superior efficacy compared with BRAF inhibitor monotherapy. CONCLUSION Targeting BRAF alterations represents a promising new therapeutic approach for a restricted subset of oncogene-addicted NSCLC. Prospect ive trials refining this strategy are ongoing. A next step will probably aim at combining BRAF inhibitors and immunotherapy or alternatively improve a multilevel mitogen-activated protein kinase (MAPK) pathway blockade by combining with ERK inhibitors.
Collapse
|
926
|
Ahmed KA, Abuodeh YA, Echevarria MI, Arrington JA, Stallworth DG, Hogue C, Naghavi AO, Kim S, Kim Y, Patel BG, Sarangkasiri S, Johnstone PAS, Sahebjam S, Khushalani NI, Forsyth PA, Harrison LB, Yu M, Etame AB, Caudell JJ. Clinical outcomes of melanoma brain metastases treated with stereotactic radiosurgery and anti-PD-1 therapy, anti-CTLA-4 therapy, BRAF/MEK inhibitors, BRAF inhibitor, or conventional chemotherapy. Ann Oncol 2016; 27:2288-2294. [PMID: 27637745 DOI: 10.1093/annonc/mdw417] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 06/30/2016] [Accepted: 08/22/2016] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND The effect of immunologic and targeted agents on intracranial response rates in patients with melanoma brain metastases (MBMs) is not yet clearly understood. This report analyzes outcomes of intact MBMs treated with single-session stereotactic radiosurgery (SRS) and anti-PD-1 therapy, anti-CTLA-4 therapy, BRAF/MEK inhibitors(i), BRAFi, or conventional chemotherapy. PATIENTS AND METHODS Patients were included if MBMs were treated with single-session SRS within 3 months of receiving systemic therapy. The primary end point of this study was distant MBM control. Secondary end points were local MBM control defined as a >20% volume increase on follow-up MRI, systemic progression-free survival, overall survival (OS) from both SRS and cranial metastases diagnosis, and neurotoxicity. Images were reviewed alongside two neuro-radiologists at our institution. RESULTS Ninety-six patients were treated to 314 MBMs over 119 SRS treatment sessions between January 2007 and August 2015. No significant differences were noted in age (P = 0.27), gender (P = 0.85), treated gross tumor volume (P = 0.26), or the diagnosis-specific graded prognostic assessment (P = 0.51) between the treatment cohorts. Twelve-month Kaplan-Meier (KM) distant MBM control rates were 38%, 21%, 20%, 8%, and 5% (P = 0.008) for SRS with anti-PD-1 therapies, anti-CTLA-4 therapy, BRAF/MEKi, BRAFi, and conventional chemotherapy, respectively. No significant differences were noted in the KM local MBM control rates among treatment groups (P = 0.25). Treatment with anti-PD-1 therapy, anti-CTLA-4 therapy, or BRAF/MEKi significantly improved OS on both univariate and multivariate analyses when compared with conventional chemotherapy. CONCLUSION In our institutional analysis of patients treated with SRS and various systemic immunologic and targeted melanoma agents, significant differences in distant MBM control and OS are noted. Prospective evaluation of the potential synergistic effect between these agents and SRS is warranted.
Collapse
Affiliation(s)
| | | | | | - J A Arrington
- Department of Radiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa
| | - D G Stallworth
- Department of Radiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa
| | - C Hogue
- Department of School of Medicine, University of Louisville, Louisville
| | | | - S Kim
- Department of Radiation Oncology
| | - Y Kim
- Department of Biostatistics, H. Lee Moffitt Cancer Center and Research Institute, Tampa
| | - B G Patel
- Department of Morsani College of Medicine, University of South Florida, Tampa
| | | | | | - S Sahebjam
- Department of Neuro-Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa
| | - N I Khushalani
- Department of Cutaneous-Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, USA
| | - P A Forsyth
- Department of Neuro-Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa
| | | | - M Yu
- Department of Radiation Oncology
| | - A B Etame
- Department of Neuro-Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa
| | | |
Collapse
|
927
|
Heppt MV, Dietrich C, Graf SA, Ruzicka T, Tietze JK, Berking C. The Systemic Management of Advanced Melanoma in 2016. Oncol Res Treat 2016; 39:635-642. [DOI: 10.1159/000448904] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 08/03/2016] [Indexed: 11/19/2022]
|
928
|
He B, Lu C, Zheng G, He X, Wang M, Chen G, Zhang G, Lu A. Combination therapeutics in complex diseases. J Cell Mol Med 2016; 20:2231-2240. [PMID: 27605177 PMCID: PMC5134672 DOI: 10.1111/jcmm.12930] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2016] [Accepted: 06/16/2016] [Indexed: 12/22/2022] Open
Abstract
The biological redundancies in molecular networks of complex diseases limit the efficacy of many single drug therapies. Combination therapeutics, as a common therapeutic method, involve pharmacological intervention using several drugs that interact with multiple targets in the molecular networks of diseases and may achieve better efficacy and/or less toxicity than monotherapy in practice. The development of combination therapeutics is complicated by several critical issues, including identifying multiple targets, targeting strategies and the drug combination. This review summarizes the current achievements in combination therapeutics, with a particular emphasis on the efforts to develop combination therapeutics for complex diseases.
Collapse
Affiliation(s)
- Bing He
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.,Institute of Integrated Bioinformedicine & Translational Science, HKBU Shenzhen Research Institute and Continuing Education, Shenzhen, China
| | - Cheng Lu
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.,Institute of Integrated Bioinformedicine & Translational Science, HKBU Shenzhen Research Institute and Continuing Education, Shenzhen, China.,Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Guang Zheng
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.,Institute of Integrated Bioinformedicine & Translational Science, HKBU Shenzhen Research Institute and Continuing Education, Shenzhen, China
| | - Xiaojuan He
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.,Institute of Integrated Bioinformedicine & Translational Science, HKBU Shenzhen Research Institute and Continuing Education, Shenzhen, China.,Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Maolin Wang
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.,Institute of Integrated Bioinformedicine & Translational Science, HKBU Shenzhen Research Institute and Continuing Education, Shenzhen, China
| | - Gao Chen
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.,Institute of Integrated Bioinformedicine & Translational Science, HKBU Shenzhen Research Institute and Continuing Education, Shenzhen, China
| | - Ge Zhang
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.,Institute of Integrated Bioinformedicine & Translational Science, HKBU Shenzhen Research Institute and Continuing Education, Shenzhen, China
| | - Aiping Lu
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.,Institute of Integrated Bioinformedicine & Translational Science, HKBU Shenzhen Research Institute and Continuing Education, Shenzhen, China.,Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
929
|
Franklin C, Livingstone E, Roesch A, Schilling B, Schadendorf D. Immunotherapy in melanoma: Recent advances and future directions. Eur J Surg Oncol 2016; 43:604-611. [PMID: 27769635 DOI: 10.1016/j.ejso.2016.07.145] [Citation(s) in RCA: 190] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2016] [Revised: 07/07/2016] [Accepted: 07/25/2016] [Indexed: 12/12/2022] Open
Abstract
Malignant melanoma contributes the majority of skin cancer related deaths and shows an increasing incidence in the past years. Despite all efforts of early diagnosis, metastatic melanoma still has a poor prognosis and remains a challenge for treating physicians. In recent years, improved knowledge of the pathophysiology and a better understanding of the role of the immune system in tumour control have led to the development and approval of several immunotherapies. Monoclonal antibodies against different immune checkpoints have been revolutionizing the treatment of metastatic and unresectable melanoma. Ipilimumab, a monoclonal antibody against the cytotoxic T-lymphocyte antigen 4 (CTLA-4) as well as nivolumab and pembrolizumab which target the programmed cell death protein 1 (PD-1) have been shown to prolong overall survival in patients with advanced melanoma. The latter substances seem to have an increased response rate and more tolerable safety profile compared to ipilimumab. The combination of a CTLA-4 and a PD-1 inhibitor seems to be superior to the monotherapies, especially in patients with PD-L1 negative tumours. Checkpoint inhibitors are currently being tested in the adjuvant setting with initial data for ipilimumab suggesting efficacy in this context. Talimogene laherparepvec (TVEC) is the first oncolytic virus approved in the therapy of metastatic melanoma offering a treatment option especially for patients with limited disease. In this review, data on these recently developed and approved immunotherapies are presented. However, further studies are necessary to determine the optimal duration, sequencing and combinations of immunotherapies to further improve the outcome of patients with advanced melanoma.
Collapse
Affiliation(s)
- C Franklin
- Department of Dermatology, Venereology and Allergology, University Hospital, University of Duisburg-Essen, Essen, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany
| | - E Livingstone
- Department of Dermatology, Venereology and Allergology, University Hospital, University of Duisburg-Essen, Essen, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany
| | - A Roesch
- Department of Dermatology, Venereology and Allergology, University Hospital, University of Duisburg-Essen, Essen, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany
| | - B Schilling
- Department of Dermatology, Venereology and Allergology, University Hospital, University of Duisburg-Essen, Essen, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany.
| | - D Schadendorf
- Department of Dermatology, Venereology and Allergology, University Hospital, University of Duisburg-Essen, Essen, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany.
| |
Collapse
|
930
|
Sammons S, Brady D, Vahdat L, Salama AK. Copper suppression as cancer therapy: the rationale for copper chelating agents in BRAFV600 mutated melanoma. Melanoma Manag 2016; 3:207-216. [PMID: 30190890 DOI: 10.2217/mmt-2015-0005] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 05/13/2016] [Indexed: 01/21/2023] Open
Abstract
The successful targeting of oncogenic BRAFV600 represents one of the landmark breakthroughs in therapy for advanced melanoma. While the initial clinical benefit can be dramatic, resistance is common due to a number of mechanisms, including MAPK pathway reactivation. Recent data have revealed a novel role for copper (Cu) in BRAF signaling with potential clinical implications. The history, preclinical data and efficacy of Cu chelating agents in cancer, specifically tetrathiomolybdate, will be reviewed with a focus on the rationale for targeting the MAPK cascade in melanoma through novel combination strategies.
Collapse
Affiliation(s)
- Sarah Sammons
- Hematology/Oncology, Department of Internal Medicine, Duke University Medical Center, 203 Research Drive, MSRB1, Room 397, Box 2639, Durham, NC 27710, USA.,Hematology/Oncology, Department of Internal Medicine, Duke University Medical Center, 203 Research Drive, MSRB1, Room 397, Box 2639, Durham, NC 27710, USA
| | - Donita Brady
- Department of Cancer Biology & Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, 421 Curie Boulevard, 612 BRBII/III, Philadelphia, PA 19104, USA.,Department of Cancer Biology & Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, 421 Curie Boulevard, 612 BRBII/III, Philadelphia, PA 19104, USA
| | - Linda Vahdat
- Division of Hematology & Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA.,Division of Hematology & Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - April Ks Salama
- Melanoma Program, Division of Medical Oncology, Department of Internal Medicine, Duke University Medical Center, 25176 Morris Bldg, DUMC 3198, Durham, NC 27710, USA.,Melanoma Program, Division of Medical Oncology, Department of Internal Medicine, Duke University Medical Center, 25176 Morris Bldg, DUMC 3198, Durham, NC 27710, USA
| |
Collapse
|
931
|
Vouk K, Benter U, Amonkar MM, Marocco A, Stapelkamp C, Pfersch S, Benjamin L. Cost and economic burden of adverse events associated with metastatic melanoma treatments in five countries. J Med Econ 2016; 19:900-12. [PMID: 27123564 DOI: 10.1080/13696998.2016.1184155] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
OBJECTIVE To estimate per-event cost and economic burden associated with managing the most common and/or severe metastatic melanoma (MM) treatment-related adverse events (AEs) in Australia, France, Germany, Italy, and the UK. METHODS AEs associated with chemotherapy (dacarbazine, paclitaxel, fotemustine), immunotherapy (ipilimumab), and targeted therapy (vemurafenib) were identified by literature review. Medical resource use data associated with managing AEs were collected through two blinded Delphi panel cycles in each of the five countries. Published costs were used to estimate per-event costs and combined with AEs incidence, treatment usage, and MM prevalence to estimate the economic burden for each country. RESULTS The costliest AEs were grade 3/4 events due to immunotherapy (Australia/France: colitis; UK: diarrhea) and chemotherapy (Germany/Italy: neutropenia/leukopenia). Treatment of AEs specific to chemotherapy (Australia/Germany/Italy/France: neutropenia/leukopenia) and targeted therapy (UK: squamous cell carcinoma) contributed heavily to country-specific economic burden. LIMITATIONS Economic burden was estimated assuming that each patient experienced an AE only once. In addition, the context of settings was heterogeneous and the number of Delphi panel experts was limited. CONCLUSIONS Management costs for MM treatment-associated AEs can be substantial. Results could be incorporated in economic models that support reimbursement dossiers. With the availability of newer treatments, establishment of a baseline measure of the economic burden of AEs will be crucial for assessing their impact on patients and regional healthcare systems.
Collapse
Affiliation(s)
| | | | | | | | | | - Sylvie Pfersch
- e Novartis Pharma S.A.S. , Rueil-Malmaison Cedex , France
| | | |
Collapse
|
932
|
Malecek MK, Robinson JK, Bilimoria K, Choi JN, Choi J, Gerami P, Kruser T, Kuzel T, Martini M, Strauss JB, Wayne J, Sosman J, Chandra S. Advancements in unresectable melanoma: a multidisciplinary perspective. Melanoma Manag 2016; 3:171-175. [PMID: 30190886 DOI: 10.2217/mmt-2016-0003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2016] [Accepted: 06/27/2016] [Indexed: 11/21/2022] Open
Affiliation(s)
- Mary-Kate Malecek
- Department of Medicine, Northwestern University Feinberg School of Medicine, 251 East Huron Street, Galter Pavilion Suite 3-150, Chicago, IL 60611, USA.,Department of Medicine, Northwestern University Feinberg School of Medicine, 251 East Huron Street, Galter Pavilion Suite 3-150, Chicago, IL 60611, USA
| | - June K Robinson
- Department of Dermatology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Department of Dermatology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Karl Bilimoria
- Division of Surgical Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Division of Surgical Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Jennifer N Choi
- Department of Dermatology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Department of Dermatology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Jaehyuk Choi
- Department of Dermatology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Department of Dermatology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Pedram Gerami
- Department of Dermatology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Department of Dermatology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Timothy Kruser
- Department of Radiation Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Department of Radiation Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Timothy Kuzel
- Division of Medical Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Division of Medical Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Mary Martini
- Department of Dermatology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Department of Dermatology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Jonathan B Strauss
- Division of Surgical Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Division of Surgical Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Jeffrey Wayne
- Division of Surgical Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Division of Surgical Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Jeffrey Sosman
- Division of Medical Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Division of Medical Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Sunandana Chandra
- Division of Medical Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Division of Medical Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
933
|
Digklia A, Michielin O. The cutting edge of metastatic melanoma therapy. Melanoma Manag 2016; 3:217-229. [PMID: 30190891 DOI: 10.2217/mmt-2016-0026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 06/16/2016] [Indexed: 11/21/2022] Open
Abstract
The past decade has witnessed impressive new developments for the treatment of melanoma. The discovery of key oncogenic driver mutations, upon which tumor establishment and progression are dependent, changed the prognosis of patients with stage IV disease. Extensive preclinical and clinical studies have shown high response rates and survival benefits over conventional chemotherapies provided by target-specific inhibitors of BRAF- or NRAS-activating mutations. Recent genomic analyses of melanoma have also given new potentially targetable driver mutations. In addition, the quickened pace of development of immune checkpoint inhibitors for the treatment of melanoma offers the unique opportunity to provide a long-term clinical benefit. In this emerging era, predictive biomarkers for the selection of patients are required to help us develop an optimal therapeutic strategy.
Collapse
Affiliation(s)
- Antonia Digklia
- Department of Oncology, Centre Hospitalier Universitaire Vaudois, Bugnon 46, 1011 Lausanne, Switzerland
| | - Olivier Michielin
- Department of Oncology, Centre Hospitalier Universitaire Vaudois, Bugnon 46, 1011 Lausanne, Switzerland
| |
Collapse
|
934
|
Gillis NK, McLeod HL. The pharmacogenomics of drug resistance to protein kinase inhibitors. Drug Resist Updat 2016; 28:28-42. [PMID: 27620953 PMCID: PMC5022787 DOI: 10.1016/j.drup.2016.06.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 06/17/2016] [Accepted: 06/29/2016] [Indexed: 01/05/2023]
Abstract
Dysregulation of growth factor cell signaling is a major driver of most human cancers. This has led to development of numerous drugs targeting protein kinases, with demonstrated efficacy in the treatment of a wide spectrum of cancers. Despite their high initial response rates and survival benefits, the majority of patients eventually develop resistance to these targeted therapies. This review article discusses examples of established mechanisms of drug resistance to anticancer therapies, including drug target mutations or gene amplifications, emergence of alternate signaling pathways, and pharmacokinetic variation. This reveals a role for pharmacogenomic analysis to identify and monitor for resistance, with possible therapeutic strategies to combat chemoresistance.
Collapse
Affiliation(s)
- Nancy K Gillis
- Eshelman School of Pharmacy, Center for Pharmacogenomics and Individualized Therapy, University of North Carolina, Chapel Hill, NC, United States; H. Lee Moffitt Cancer Center and Research Institute, DeBartolo Family Personalized Medicine Institute, Tampa, FL, United States
| | - Howard L McLeod
- H. Lee Moffitt Cancer Center and Research Institute, DeBartolo Family Personalized Medicine Institute, Tampa, FL, United States; Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
935
|
Ascierto PA, McArthur GA, Dréno B, Atkinson V, Liszkay G, Di Giacomo AM, Mandalà M, Demidov L, Stroyakovskiy D, Thomas L, de la Cruz-Merino L, Dutriaux C, Garbe C, Yan Y, Wongchenko M, Chang I, Hsu JJ, Koralek DO, Rooney I, Ribas A, Larkin J. Cobimetinib combined with vemurafenib in advanced BRAFV600-mutant melanoma (coBRIM): updated efficacy results from a randomised, double-blind, phase 3 trial. Lancet Oncol 2016; 17:1248-60. [DOI: 10.1016/s1470-2045(16)30122-x] [Citation(s) in RCA: 726] [Impact Index Per Article: 80.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2016] [Revised: 04/26/2016] [Accepted: 04/26/2016] [Indexed: 11/27/2022]
|
936
|
Kim YH. Dual inhibition of BRAF and MEK in BRAF-mutated metastatic non-small cell lung cancer. J Thorac Dis 2016; 8:2369-2371. [PMID: 27746978 DOI: 10.21037/jtd.2016.09.16] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Young Hak Kim
- Department of Respiratory Medicine, Kyoto University Hospital, Kyoto, Japan
| |
Collapse
|
937
|
Noeparast A, Teugels E, Giron P, Verschelden G, De Brakeleer S, Decoster L, De Grève J. Non-V600 BRAF mutations recurrently found in lung cancer predict sensitivity to the combination of Trametinib and Dabrafenib. Oncotarget 2016; 8:60094-60108. [PMID: 28947956 PMCID: PMC5601124 DOI: 10.18632/oncotarget.11635] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 06/09/2016] [Indexed: 12/12/2022] Open
Abstract
Approximately half of BRAF-mutated Non-small cell lung cancers (NSCLCs) harbor a non-V600 BRAF mutation, accounting for ∼40,000 annual deaths worldwide. Recent studies have revealed the benefits of combined targeted therapy with a RAF-inhibitor (Dabrafenib) and a MEK-inhibitor (Trametinib) in treating V600 BRAF mutant cancers, including NSCLC. In contrast, sensitivity of non-V600 BRAF mutations to these inhibitors is not documented. Non-V600 mutations can either increase or impair BRAF kinase activity. However, impaired BRAF kinases can still activate the ERK pathway in a CRAF-dependent manner. Herein, beyond describing a cohort of BRAF mutant NSCLC patients and functionally analyzing 13 tumor-derived BRAF mutations, we demonstrate that both types of non-V600 BRAF mutations can be sensitive to clinically relevant doses of Dabrafenib and Trametinib in HEK293T cells, in lung epithelial cellular model (BEAS-2B) and in human cancer cell lines harboring non-V600 BRAF mutations. ERK activity induced by both types of these mutations is further reduced by combinatorial drug treatment. Moreover, the combination leads to more prolonged ERK inhibition and has anti-proliferative and pro-apoptotic effects in cells harboring both types of non-V600 BRAF mutations. This study provides a basis for the clinical exploration of non-V600 BRAF mutant lung cancers upon treatment with Trametinib and Dabrafenib.
Collapse
Affiliation(s)
- Amir Noeparast
- Laboratory of Molecular Oncology and Department of Medical Oncology, Oncologisch Centrum, UZ Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Erik Teugels
- Laboratory of Molecular Oncology and Department of Medical Oncology, Oncologisch Centrum, UZ Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Philippe Giron
- Laboratory of Molecular Oncology and Department of Medical Oncology, Oncologisch Centrum, UZ Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Gil Verschelden
- Laboratory of Molecular Oncology and Department of Medical Oncology, Oncologisch Centrum, UZ Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Sylvia De Brakeleer
- Laboratory of Molecular Oncology and Department of Medical Oncology, Oncologisch Centrum, UZ Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Lore Decoster
- Laboratory of Molecular Oncology and Department of Medical Oncology, Oncologisch Centrum, UZ Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Jacques De Grève
- Laboratory of Molecular Oncology and Department of Medical Oncology, Oncologisch Centrum, UZ Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
938
|
Heo JR, Kim NH, Cho J, Choi KC. Current treatments for advanced melanoma and introduction of a promising novel gene therapy for melanoma (Review). Oncol Rep 2016; 36:1779-86. [PMID: 27573048 DOI: 10.3892/or.2016.5032] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 05/24/2016] [Indexed: 11/06/2022] Open
Abstract
Metastatic melanoma is a fatal form of skin cancer that has a tendency to proliferate more rapidly than any other solid tumor. Since 2010, treatment options for metastatic melanoma have been developed including chemotherapies, checkpoint inhibition immunotherapies, e.g., anti‑cytotoxic T‑lymphocyte antigen‑4 (CTLA‑4) and anti‑programmed death‑1 (PD‑1), and molecular-targeted therapies, e.g., BRAF and MEK inhibitors. These treatments have shown not only high response rates yet also side‑effects and limitations. Notwithstanding its limitations, stem cell therapy has emerged as a new auspicious therapy for various tumor types. Since stem cells possess the ability to serve as a novel vehicle for delivering therapeutic or suicide genes to primary or metastatic cancer sites, these cells can function as part of gene‑directed enzyme prodrug therapy (GDEPT). This review focuses on introducing engineered neural stem cells (NSCs), which have tumor‑tropic behavior that allows NSCs to selectively approach primary and invasive tumor foci, as a potential gene therapy for melanoma. Therapy using engineered NSCs with cytotoxic agents resulted in markedly reduced tumor volumes and significantly prolonged survival rates in preclinical models of various tumor types. This review elucidates current treatment options for metastatic melanoma and introduces a promising NSC therapy.
Collapse
Affiliation(s)
- Jae-Rim Heo
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Nam-Hyung Kim
- Department of Animal Science, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Jaejin Cho
- Department of Dental Regenerative Biotechnology, Seoul National University, Seoul, Republic of Korea
| | - Kyung-Chul Choi
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| |
Collapse
|
939
|
Amin A, Lawson DH, Salama AKS, Koon HB, Guthrie T, Thomas SS, O'Day SJ, Shaheen MF, Zhang B, Francis S, Hodi FS. Phase II study of vemurafenib followed by ipilimumab in patients with previously untreated BRAF-mutated metastatic melanoma. J Immunother Cancer 2016; 4:44. [PMID: 27532019 PMCID: PMC4986368 DOI: 10.1186/s40425-016-0148-7] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 07/11/2016] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Ipilimumab (IPI), an anti-CTLA-4 antibody, and vemurafenib (VEM), a BRAF inhibitor, have distinct mechanisms of action and shared toxicities (e.g., skin, gastrointestinal [GI] and hepatobiliary disorders) that may preclude concomitant administration. Concurrent administration of IPI and VEM previously showed significant dose-limiting hepatotoxicity in advanced melanoma. This single-arm, open-label, phase II study evaluated a sequencing strategy with these two agents in previously untreated patients with BRAF-mutated advanced melanoma. METHODS This study was divided into two parts. During Part 1 (VEM1-IPI), patients received VEM 960 mg twice daily for 6 weeks followed by IPI 10 mg/kg every 3 weeks for 4 doses (induction), then every 12 weeks (maintenance) beginning at week 24 until disease progression or unacceptable toxicity. During Part 2 (VEM2), patients who progressed after IPI received VEM at their previously tolerated dose. The primary objective was to estimate the incidence of grade 3/4 drug-related skin adverse events (AEs) during VEM1-IPI. RESULTS All patients who were initially treated with VEM (n = 46) received IPI induction therapy; 8 received IPI maintenance and 19 were treated during VEM2. During VEM1-IPI, the incidence of grade 3/4 drug-related AEs associated with the skin, GI tract, and hepatobiliary system was 32.6 %, 21.7 %, and 4.3 %, respectively. There were no drug-related deaths. At a median follow-up of 15.3 months, median overall survival was 18.5 months. Median progression-free survival was 4.5 months. CONCLUSIONS VEM (960 mg twice daily for 6 weeks) followed by IPI 10 mg/kg has a manageable safety profile. The benefits/risks of BRAF inhibitors followed by immunotherapy should be evaluated further in light of continuing developments in treatment options for metastatic melanoma. TRIAL REGISTRATION ClinicalTrials.gov identifier: NCT01673854 (CA184-240) Registered 24 August 2012.
Collapse
Affiliation(s)
- Asim Amin
- Levine Cancer Institute, Carolinas Healthcare System, Medical Oncology, 1021 Morehead Medical Drive, Charlotte, NC 28204 USA
| | - David H Lawson
- Winship Cancer Institute of Emory University, Atlanta, GA USA
| | | | - Henry B Koon
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH USA
| | | | | | - Steven J O'Day
- John Wayne Cancer Institute at Providence Saint John's Health Center, Santa Monica, CA USA
| | | | - Bin Zhang
- Bristol-Myers Squibb, Princeton, NJ USA ; Current Address: KBP BioSciences, Princeton, NJ USA
| | | | | |
Collapse
|
940
|
Mateus C, Libenciuc C, Robert C. [Not Available]. Bull Cancer 2016; 103:S4-S11. [PMID: 27494973 DOI: 10.1016/s0007-4551(16)30140-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
ANTI-PD1 ROLE IN TREATMENT OF CUTANEOUS MELANOMA: The treatment of metastatic melanoma dramatically changed over the last years. Two therapeutic revolutions emerged in parallel, targeted anti-BRAF and anti-MEK therapies, for patients BRAFV600 mutated and immunotherapy with immune checkpoint blockers using anti-CTLA-4 then anti-PD1 monoclonal antibodies. Indeed, melanoma immunotherapy was a golden objective for many years but in spite of important efforts using cytokines (interferon, interleukin) and different vaccine approaches no objective improvement of patients 'prognosis was obtained. Ipilimumab, authorized in 2011, was the first drug which showed a benefit of overall survival in patients with metastatic melanoma in spite a low response rate (10-15) and the occurrence of about 25% of serious toxicity. Anti-PD1 appear as a new generation of immune checkpoint blockade with response rates between 30 to 40% of the patients, a proven overall survival benefit as compared with chemotherapy or ipilimumab and less toxicity than ipilimumab. Two molecules, pembrolizumab and nivolumab were recently approved in monotherapy, for metastatic melanoma. Several questions remain unresolved: the respective indications of anti-PD1 and targeted therapies in first line therapy in patients with BRAF mutant melanoma, the benefit of combining immunotherapy with radiotherapy or with targeted therapies, the optimal treatment duration, and the benefit of the anti-PD1 in the adjuvant setting. The combination of ipilimumab and nivolumab, recently approved by the FDA but not yet in Europ, shows an improvement of the objective response rates (50-57%) and progression free survival compared with nivolumab but is associated with an higer incidence of serious adverse events (more than 50%).
Collapse
Affiliation(s)
- Christine Mateus
- Service de dermatologie, Gustave Roussy, Université Paris-Saclay, Département de Médecine Oncologique, Villejuif, F-94805, France.
| | - Cristina Libenciuc
- Service de dermatologie, Gustave Roussy, Université Paris-Saclay, Département de Médecine Oncologique, Villejuif, F-94805, France
| | - Caroline Robert
- Service de dermatologie, Gustave Roussy, Université Paris-Saclay, Département de Médecine Oncologique, Villejuif, F-94805, France
| |
Collapse
|
941
|
Abstract
With the rapid succession of new effective agents for melanoma in the recent years, the paradigm for treatment of metastatic melanoma is changing. The success of combining multiple effective agents compared with outcomes of monotherapy also brings increasing complexity in the treatment algorithm for various subsets of metastatic melanoma patients. We reviewed the recent reports on novel melanoma therapy to shed light on rational decision-making in treating these patients.
Collapse
|
942
|
Urner-Bloch U, Urner M, Jaberg-Bentele N, Frauchiger AL, Dummer R, Goldinger SM. MEK inhibitor-associated retinopathy (MEKAR) in metastatic melanoma: Long-term ophthalmic effects. Eur J Cancer 2016; 65:130-8. [PMID: 27497344 DOI: 10.1016/j.ejca.2016.06.018] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 06/16/2016] [Indexed: 11/17/2022]
Abstract
BACKGROUND Mitogen-activated protein kinase kinase (MEK) inhibitors have aroused considerable interest in oncology. Activity has been demonstrated in various types of cancer, especially melanoma. MEK inhibitors induce a transient retinopathy, considered to be a class effect. At present, only sparse data are available on retinal effects with long-term MEK inhibition. PATIENTS AND METHODS In this prospective, observational study, patients with advanced melanoma participating in different phase 1/2 or phase 3 clinical trials were treated with the MEK inhibitor binimetinib, with a v-Raf murine sarcoma viral oncogene homolog B (BRAF) inhibitor, or with combination therapy. They underwent regular ophthalmological examinations including determination of visual function, biomicroscopy, dilated fundoscopy and optical coherence tomography (OCT) for a period of up to 2 years. Retinopathy was diagnosed on defined OCT criteria. RESULTS Sixty-two patients were investigated between 1st October 2011 and 31st July 2015: 13 were treated with the MEK inhibitor binimetinib alone, 10 with a selective BRAF inhibitor, and 39 with combination therapy. In 92% of patients on monotherapy and 100% of those on combination treatment, binimetinib caused dose-related lesions with serous neuroretinal detachments and oedema, strongly dependent on the time after medication. With continued treatment, retinal volume and thickness decreased to levels below baseline, without any apparent functional deficits or changes in structural integrity. CONCLUSIONS Binimetinib induces a specific retinopathy with daily fluctuations depending on the time interval after medication. The retinopathy partially recovers, but can still be detected many months later. Retinal thinning, possible first signs of retinal atrophy have been observed after long-term treatment, but, so far, without functional relevance.
Collapse
Affiliation(s)
- U Urner-Bloch
- Private Ophthalmic Practice in Cooperation with the Skin Cancer Unit, University Hospital of Zurich, Zurich, Switzerland
| | - M Urner
- Medical Intensive Care Unit, University Hospital Zurich, Zurich, Switzerland; Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - N Jaberg-Bentele
- Department of Dermatology, University Hospital of Zurich, Zurich, Switzerland
| | - A L Frauchiger
- Department of Dermatology, University Hospital of Zurich, Zurich, Switzerland
| | - R Dummer
- Department of Dermatology, University Hospital of Zurich, Zurich, Switzerland.
| | - S M Goldinger
- Department of Dermatology, University Hospital of Zurich, Zurich, Switzerland
| |
Collapse
|
943
|
Wang DY, Johnson DB. Update on immune therapy in melanoma. Expert Opin Orphan Drugs 2016. [DOI: 10.1080/21678707.2016.1194752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
944
|
Initial experience with combined BRAF and MEK inhibition with stereotactic radiosurgery for BRAF mutant melanoma brain metastases. Melanoma Res 2016; 26:382-6. [DOI: 10.1097/cmr.0000000000000250] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
945
|
Simberg-Danell C, Lyth J, Månsson-Brahme E, Frohm-Nilsson M, Carstensen J, Hansson J, Eriksson H. Prognostic factors and disease-specific survival among immigrants diagnosed with cutaneous malignant melanoma in Sweden. Int J Cancer 2016; 139:543-53. [PMID: 27004457 DOI: 10.1002/ijc.30103] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2015] [Revised: 01/30/2016] [Accepted: 02/19/2016] [Indexed: 02/03/2023]
Abstract
Little is known about cutaneous malignant melanoma (CMM) among immigrants in Europe. We aimed to investigate clinical characteristics and disease-specific survival among first- and second-generation immigrants in Sweden. This nationwide population-based study included 27,235 patients from the Swedish Melanoma Register diagnosed with primary invasive CMM, 1990-2007. Data were linked to nationwide, population-based registers followed up through 2013. Logistic regression and Cox regression models were used to determine the association between immigrant status, stage and CMM prognosis, respectively. After adjustments for confounders, first generation immigrants from Southern Europe were associated with significantly more advanced stages of disease compared to Swedish-born patients [Stage II vs. I: Odds ratio (OR) = 2.37, 95% CI = 1.61-3.50. Stage III-IV vs I: OR = 2.40, 95% CI = 1.08-5.37]. The ORs of stage II-IV versus stage I disease were increased among men (OR = 1.9; 95% CI = 1.1-3.3; p = 0.020), and women (OR = 4.8; 95% CI = 2.6-9.1; p < 0.001) in a subgroup of immigrants from former Yugoslavia compared to Swedish-born patients. The CMM-specific survival was significantly decreased among women from former Yugoslavia versus Swedish-born women [hazard ratio (HR)=2.2; 95% CI = 1.1-4.2; p = 0.043]. After additional adjustments including stage, the survival difference was no longer significant. No survival difference between the second generation immigrant group and Swedish-born patients were observed. In conclusion, a worse CMM-specific survival in women from former Yugoslavia was associated with more advanced stages of CMM at diagnosis. Secondary prevention efforts focusing on specific groups may be needed to further improve the CMM prognosis.
Collapse
Affiliation(s)
- Caroline Simberg-Danell
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, SE, 171 76, Sweden
- Department of Dermatology and Venereology, Södersjukhuset, Stockholm, SE, 118 83, Sweden
| | - Johan Lyth
- Unit of Research and Development in Local Health Care, County of Östergötland, Linköping, Sweden
| | - Eva Månsson-Brahme
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, SE, 171 76, Sweden
- Department of Oncology and Pathology, Karolinska University Hospital, Stockholm, SE, 171 76, Sweden
| | - Margareta Frohm-Nilsson
- Department of Medicine, Unit of Dermatology, Karolinska University Hospital, Stockholm, SE, 171 76, Sweden
| | - John Carstensen
- Division of Health Care Analysis, Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
| | - Johan Hansson
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, SE, 171 76, Sweden
- Department of Oncology and Pathology, Karolinska University Hospital, Stockholm, SE, 171 76, Sweden
| | - Hanna Eriksson
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, SE, 171 76, Sweden
- Department of Oncology and Pathology, Karolinska University Hospital, Stockholm, SE, 171 76, Sweden
| |
Collapse
|
946
|
Cabanillas ME, Busaidy NL, Khan SA, Gunn GB, Dadu R, Rao SN, Waguespack SG. Molecular diagnostics and anaplastic thyroid carcinoma: the time has come to harvest the high hanging fruit. INTERNATIONAL JOURNAL OF ENDOCRINE ONCOLOGY 2016. [DOI: 10.2217/ije-2016-0007] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Targeted therapies have played a major role in cancer therapeutics, starting with the discovery of a drug against BCR–ABL rearrangements in chronic myelogenous leukemia. This led to the first approval of a targeted agent in cancer and since, many others have followed. Anaplastic thyroid cancer (ATC) is an aggressive carcinoma with few curative options. Although previous cytotoxic chemotherapy and kinase inhibitor therapies have not proven efficacious in ATC, some of the newer drugs appear to be promising. A case report and a comprehensive review of the current standard of care, genetics, modern therapeutic drugs and clinical trials are presented, in order to outline where we currently stand and where the future lies in the quest for a cure for ATC.
Collapse
Affiliation(s)
- Maria E Cabanillas
- Department of Endocrine Neoplasia & Hormonal Disorders, The University of Texas MD Anderson Cancer Center; Houston, TX, USA
| | - Naifa L Busaidy
- Department of Endocrine Neoplasia & Hormonal Disorders, The University of Texas MD Anderson Cancer Center; Houston, TX, USA
| | - Saad A Khan
- The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - G Brandon Gunn
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center; Houston, TX, USA
| | - Ramona Dadu
- Department of Endocrine Neoplasia & Hormonal Disorders, The University of Texas MD Anderson Cancer Center; Houston, TX, USA
| | - Sarika N Rao
- Department of Endocrine Neoplasia & Hormonal Disorders, The University of Texas MD Anderson Cancer Center; Houston, TX, USA
| | - Steven G Waguespack
- Department of Endocrine Neoplasia & Hormonal Disorders, The University of Texas MD Anderson Cancer Center; Houston, TX, USA
| |
Collapse
|
947
|
Combined Therapy with Dabrafenib and Trametinib in BRAF-Mutated Metastatic Melanoma in a Real-Life Setting: The INT Milan Experience. TUMORI JOURNAL 2016; 102:501-507. [DOI: 10.5301/tj.5000539] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/12/2016] [Indexed: 11/20/2022]
Abstract
Purpose Combination therapy with dabrafenib and trametinib is safer and more effective than BRAF inhibitor-based monotherapy for metastatic melanoma. Methods We retrospectively analyzed BRAF-mutated metastatic melanoma patients treated at our institution with daily oral dabrafenib 300 mg and trametinib 2 mg from November 2013 to April 2016. This clinical record included both untreated and previously treated stage IV melanomas. Physical examination and laboratory examinations were performed monthly and disease re-evaluations were performed every 3 months. Results A total of 48 patients (24 male, 24 female) with BRAF-mutated metastatic melanoma received dabrafenib and trametinib; median age was 48 years (range 23-75). Median follow-up was 362.5 days (range 72-879). Best overall response rate consisted of 6.2% (3 patients) complete response, 64.6% (31) partial response, and 25% ( 12 ) stable disease; median time to best response was 11 weeks (range 5.7-125.5). Progression of disease was seen in 19 patients (39.6%), with median time to progression (TTP) of 26 weeks (range 8-54). A total of 15 patients (31.2%) died due to progression of disease. Median progression-free survival and median overall survival were not reached. To date, 30 patients (62.5%) are still under treatment. A total of 27 (56.2%) patients had at least one adverse event (AE); grade 3-4 AEs were seen in 4 cases (8.3%). The main toxicities were fever (25%), skin rash (14.6%), arthralgias (10.4%), and aspartate aminotransferase/alanine aminotransferase increase (8.3%). Treatment dose was reduced in 7 subjects (14.6%), with only one case of discontinuation due to AE. Conclusions Our data, using combined targeted therapy, are in line with the scientific literature in terms of both safety and effectiveness in a real-life setting.
Collapse
|
948
|
Abstract
INTRODUCTION The treatment of melanoma is evolving rapidly over the past few years. Patients with BRAFv600 mutations can be treated with a combination of a BRAF-inhibitor and an MEK-inhibitor. Patients with BRAF wild-type tumors and BRAFv600 mutated tumors can be treated with immunotherapy i.e. check point inhibitors. AREAS COVERED We conducted a comprehensive review of the literature on the efficacy and predictive markers, safety, and pharmacoeconomics of ipilimumab in melanoma Expert commentary: Ipilimumab was the first check point inhibitor reaching the clinic, gaining FDA and EMA approval for metastatic melanoma in 2011. Ipilimumab was also approved by FDA in the adjuvant setting for patients with high risk, stage III melanoma. The anti-PD1 directed antibodies pembrolizumab and nivolumab are superior to single agent ipilimumab, which is no longer considered the standard first line treatment in metastatic melanoma. The addition ipilimumab to nivolumab is associated with a higher response rate and a better PFS, particularly in patients with PD-L1 negative tumors, albeit at the cost of a steep increase in grade 3-4 adverse event rate. Definitive survival data on this combination are pending and the selection of patients potentially requiring the combination and its pharmacoeconomic implications are to be elucidated.
Collapse
Affiliation(s)
- Pol Specenier
- a Oncology , Universitair Ziekenhuis Antwerpen , Edegem , Belgium
| |
Collapse
|
949
|
Jensen IS, Zacherle E, Blanchette CM, Zhang J, Yin W. Evaluating cost benefits of combination therapies for advanced melanoma. Drugs Context 2016; 5:212297. [PMID: 27540409 PMCID: PMC4974051 DOI: 10.7573/dic.212297] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Although a number of monoimmunotherapies and targeted therapies are available to treat BRAF+ advanced melanoma, response rates remain relatively low in the range of 22-53% with progression-free survival (PFS) in the range of 4.8-8.8 months. Recently, combination targeted therapies have improved response rates to about 66-69%, PFS to 11.0-12.6 months and overall survival (OS) to 25.1-25.6 months. While combination immunotherapies have improved response rates of 67 compared with 19-29% with monotherapies and improved PFS of 11.7 compared with 4.4-5.8 months with monotherapies, the OS benefit is yet to be established in phase 3 trials. As healthcare costs continue to rise, US payers have a predominant interest in assessing the value of available treatments. Therefore, a cost-benefit model was developed to evaluate the value of treating BRAF+ advanced melanoma with two combination therapies: nivolumab + ipilimumab (N+I) and dabrafenib + trametinib (D+T). SCOPE The model was used to estimate total costs, total costs by expenditure category, cost per month of PFS and cost per responder for the payer, and societal perspectives of treating advanced melanoma patients with the BRAF V600 mutation using combination targeted therapy (D+T) or combination immunotherapy (N+I). The model followed patients from initiation of treatment to the point of progression or death. Deterministic and probabilistic sensitivity analyses were conducted to evaluate the robustness of the results and to understand the dispersion of simulated results. FINDINGS Based on a hypothetical payer with one million covered lives, it was expected that fourteen metastatic melanoma patients with the BRAF V600 mutation would be treated each year. Cost-benefit with N+I and D+T was simulated from the payer perspective. The cost per month of PFS for N+I was $22,162, while that for D+T was $17,716 (-$4,446 cost difference); the cost per responder for N+I was $388,746 and that for D+T was $282,429 (-$106,316 cost difference). The cost per month of PFS and per responder from the societal perspective resembled the patterns observed from the payer's perspective: the cost per month of PFS for N+I was $22,843, while that for D+T was $18,283 (-$4,560 cost difference). The cost per responder for N+I was $400,695 and that for D+T was $291,473 (-$109,222 cost difference). The totals of travel and treatment time for N+I and D+T were 58 hours and 3.9 hours per patient, respectively, of which total infusion time for N+I accounted for a majority - 59% - of the 58 hours. Sensitivity analyses indicated that results were most sensitive to model inputs for median PFS, body weight, and drug cost. Moreover, D+T is likely associated with a lower cost per month of PFS and cost per responder than N+I, except at low body weights (less than 57 kg). CONCLUSION The model presented in this study was used to analyze the clinical and economic benefit of using combination therapies in advanced melanoma patients with the BRAF V600 mutation. This analysis suggests D+T therapy is associated with less patient time and lower costs relative to N+I to gain similar PFS and overall response rate (ORR) benefits.
Collapse
Affiliation(s)
- Ivar S. Jensen
- Precision Health Economics, 101 Tremont St. Suite 400, Boston, MA 02108, USA
| | - Emily Zacherle
- Precision Health Economics, 209 Delburg St. Suite 106, Davidson, NC 28036, USA
- University of North Carolina at Charlotte, 9201 University City Blvd,Charlotte, NC 28223, USA
| | - Christopher M. Blanchette
- Precision Health Economics, 209 Delburg St. Suite 106, Davidson, NC 28036, USA
- University of North Carolina at Charlotte, 9201 University City Blvd,Charlotte, NC 28223, USA
| | - Jie Zhang
- Novartis Pharmaceuticals Corporation, One Health Plaza, BLDG 315, 5550B, East Hanover, NJ 07936, USA
| | - Wes Yin
- University of California at Los Angelos, 11100 Santa Monica Blvd. Suite 500, Los Angelos, CA 90025, USA
| |
Collapse
|
950
|
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: 1.1] [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
|