1701
|
Albertini MR, Ranheim EA, Zuleger CL, Sondel PM, Hank JA, Bridges A, Newton MA, McFarland T, Collins J, Clements E, Henry MB, Neuman HB, Weber S, Whalen G, Galili U. Phase I study to evaluate toxicity and feasibility of intratumoral injection of α-gal glycolipids in patients with advanced melanoma. Cancer Immunol Immunother 2016; 65:897-907. [PMID: 27207605 DOI: 10.1007/s00262-016-1846-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 05/08/2016] [Indexed: 12/14/2022]
Abstract
Effective uptake of tumor cell-derived antigens by antigen-presenting cells is achieved pre-clinically by in situ labeling of tumor with α-gal glycolipids that bind the naturally occurring anti-Gal antibody. We evaluated toxicity and feasibility of intratumoral injections of α-gal glycolipids as an autologous tumor antigen-targeted immunotherapy in melanoma patients (pts). Pts with unresectable metastatic melanoma, at least one cutaneous, subcutaneous, or palpable lymph node metastasis, and serum anti-Gal titer ≥1:50 were eligible for two intratumoral α-gal glycolipid injections given 4 weeks apart (cohort I: 0.1 mg/injection; cohort II: 1.0 mg/injection; cohort III: 10 mg/injection). Monitoring included blood for clinical, autoimmune, and immunological analyses and core tumor biopsies. Treatment outcome was determined 8 weeks after the first α-gal glycolipid injection. Nine pts received two intratumoral injections of α-gal glycolipids (3 pts/cohort). Injection-site toxicity was mild, and no systemic toxicity or autoimmunity could be attributed to the therapy. Two pts had stable disease by RECIST lasting 8 and 7 months. Tumor nodule biopsies revealed minimal to no change in inflammatory infiltrate between pre- and post-treatment biopsies except for 1 pt (cohort III) with a post-treatment inflammatory infiltrate. Two and four weeks post-injection, treated nodules in 5 of 9 pts exhibited tumor cell necrosis without neutrophilic or lymphocytic inflammatory response. Non-treated tumor nodules in 2 of 4 evaluable pts also showed necrosis. Repeated intratumoral injections of α-gal glycolipids are well tolerated, and tumor necrosis was seen in some tumor nodule biopsies after tumor injection with α-gal glycolipids.
Collapse
Affiliation(s)
- Mark R Albertini
- University of Wisconsin Carbone Cancer Center, Madison, WI, USA.
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.
- Medical Service, William S. Middleton Memorial Veterans Hospital, Madison, WI, USA.
- University of Wisconsin Clinical Sciences Center, Room K6/530, 600 Highland Avenue, Madison, WI, 53792, USA.
| | - Erik A Ranheim
- University of Wisconsin Carbone Cancer Center, Madison, WI, USA
- Department of Pathology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Cindy L Zuleger
- University of Wisconsin Carbone Cancer Center, Madison, WI, USA
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Paul M Sondel
- University of Wisconsin Carbone Cancer Center, Madison, WI, USA
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Jacquelyn A Hank
- University of Wisconsin Carbone Cancer Center, Madison, WI, USA
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Alan Bridges
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Medical Service, William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Michael A Newton
- University of Wisconsin Carbone Cancer Center, Madison, WI, USA
- Department of Biostatistics and Medical Informatics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Thomas McFarland
- University of Wisconsin Carbone Cancer Center, Madison, WI, USA
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | | | - Erin Clements
- University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - Mary Beth Henry
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Heather B Neuman
- University of Wisconsin Carbone Cancer Center, Madison, WI, USA
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Sharon Weber
- University of Wisconsin Carbone Cancer Center, Madison, WI, USA
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Giles Whalen
- Department of Surgery, University of Massachusetts Medical School, Worcester, MA, USA
| | - Uri Galili
- Department of Surgery, University of Massachusetts Medical School, Worcester, MA, USA
| |
Collapse
|
1702
|
Bui L, Hendricks A, Wright J, Chuong CJ, Davé D, Bachoo R, Kim YT. Brain Tumor Genetic Modification Yields Increased Resistance to Paclitaxel in Physical Confinement. Sci Rep 2016; 6:26134. [PMID: 27184621 PMCID: PMC4869028 DOI: 10.1038/srep26134] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Accepted: 04/27/2016] [Indexed: 01/13/2023] Open
Abstract
Brain tumor cells remain highly resistant to radiation and chemotherapy, particularly malignant and secondary cancers. In this study, we utilized microchannel devices to examine the effect of a confined environment on the viability and drug resistance of the following brain cancer cell lines: primary cancers (glioblastoma multiforme and neuroblastoma), human brain cancer cell lines (D54 and D54-EGFRvIII), and genetically modified mouse astrocytes (wild type, p53-/-, p53-/- PTEN-/-, p53-/- Braf, and p53-/- PTEN-/- Braf). We found that loss of PTEN combined with Braf activation resulted in higher viability in narrow microchannels. In addition, Braf conferred increased resistance to the microtubule-stabilizing drug Taxol in narrow confinement. Similarly, survival of D54-EGFRvIII cells was unaffected following treatment with Taxol, whereas the viability of D54 cells was reduced by 75% under these conditions. Taken together, our data suggests key targets for anticancer drugs based on cellular genotypes and their specific survival phenotypes during confined migration.
Collapse
Affiliation(s)
- Loan Bui
- Department of Bioengineering, University of Texas at Arlington, TX, USA
| | - Alissa Hendricks
- Department of Bioengineering, University of Texas at Arlington, TX, USA
| | - Jamie Wright
- Department of Bioengineering, University of Texas at Arlington, TX, USA
| | - Cheng-Jen Chuong
- Department of Bioengineering, University of Texas at Arlington, TX, USA
| | - Digant Davé
- Department of Bioengineering, University of Texas at Arlington, TX, USA.,Advanced Imaging Research Center, UT Southwestern Medical Center, TX, USA
| | - Robert Bachoo
- Department of Neurology and Neurotherapeutics, UT Southwestern Medical Center, TX, USA
| | - Young-Tae Kim
- Department of Bioengineering, University of Texas at Arlington, TX, USA.,Department of Urology, UT Southwestern Medical Center, TX, USA
| |
Collapse
|
1703
|
[Systemic treatment of inoperable metastasized malignant melanoma]. Hautarzt 2016; 67:529-35. [PMID: 27164828 DOI: 10.1007/s00105-016-3795-1] [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
BACKGROUND The medical therapy of inoperable malignant melanoma has changed dramatically over the last few years. OBJECTIVES The purpose of this article is to summarize the current state of systemic medical treatment of malignant melanoma. MATERIALS AND METHODS Clinical studies and guidelines in the therapy of malignant melanoma are reviewed. RESULTS Medical therapy of inoperable melanoma changed due to developments in immunotherapies (checkpoint inhibitors) and molecular-targeted therapies (BRAF and MEK inhibitors). Checkpoint inhibitors are antibodies administered as infusions every 2-3 weeks, blocking the checkpoints PD-1 or CTLA-4, thus, preventing downregulation of the immune system. BRAF and MEK inhibitors are small molecules, they are given orally and block a certain signaling pathway in tumor cells. The activation of this pathway has to be demonstrated by molecular analysis of tumor tissue first. This strategy is currently registered for 40-50 % of melanomas harboring a BRAF V600 mutation, while the combination of a BRAF plus MEK inhibitor has been proven more efficient than a BRAF inhibitor alone. DISCUSSION A fascinating development has started in the melanoma field due to immunotherapeutic and molecular-targeted treatment strategies. The continuation of this development needs further clinical and translational studies. This includes particular clinical studies with the new substances in the adjuvant situation, and sequences and combinations in the metastatic setting. Translational studies are needed to develop biomarkers for response and side effects.
Collapse
|
1704
|
Eroglu Z, Smalley KSM, Sondak VK. Improving patient outcomes to targeted therapies in melanoma. Expert Rev Anticancer Ther 2016; 16:633-41. [PMID: 27137746 DOI: 10.1080/14737140.2016.1178575] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
INTRODUCTION The arrival of targeted therapies has led to significant improvements in clinical outcomes for patients with BRAFV600 mutated advanced melanoma over the past five years. AREAS COVERED In several clinical trials, BRAF and MEK inhibitors have shown improvement in progression free and overall survival, along with much higher tumor response rates in comparison to chemotherapy, with the combination of these drugs superior to monotherapy. These agents are also being tested in earlier-stage patients, in addition to alternative dosing regimens and in combinations with other therapeutics. Efforts are also ongoing to expand the success found with targeted therapies to other subtypes of melanoma, including NRAS and c-kit mutated melanomas, uveal melanomas, and BRAF/NRAS wild type melanomas. Expert Commentary: We aim to provide an overview of clinical outcomes with targeted therapies in melanoma patients.
Collapse
Affiliation(s)
- Zeynep Eroglu
- a The Department of Cutaneous Oncology , The Moffitt Cancer Center & Research Institute , Tampa , FL , USA
| | - Keiran S M Smalley
- a The Department of Cutaneous Oncology , The Moffitt Cancer Center & Research Institute , Tampa , FL , USA.,b The Department of Tumor Biology , The Moffitt Cancer Center & Research Institute , Tampa , FL , USA
| | - Vernon K Sondak
- a The Department of Cutaneous Oncology , The Moffitt Cancer Center & Research Institute , Tampa , FL , USA
| |
Collapse
|
1705
|
Levesque MP, Dummer R, Beerenwinkel N. Perturbing resistance: a network perspective. Pigment Cell Melanoma Res 2016; 29:5-7. [PMID: 26471867 DOI: 10.1111/pcmr.12431] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
1706
|
Martens A, Wistuba-Hamprecht K, Yuan J, Postow MA, Wong P, Capone M, Madonna G, Khammari A, Schilling B, Sucker A, Schadendorf D, Martus P, Dreno B, Ascierto PA, Wolchok JD, Pawelec G, Garbe C, Weide B. Increases in Absolute Lymphocytes and Circulating CD4+ and CD8+ T Cells Are Associated with Positive Clinical Outcome of Melanoma Patients Treated with Ipilimumab. Clin Cancer Res 2016; 22:4848-4858. [PMID: 27169993 DOI: 10.1158/1078-0432.ccr-16-0249] [Citation(s) in RCA: 139] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 04/26/2016] [Indexed: 01/05/2023]
Abstract
PURPOSE To investigate changes of peripheral blood biomarkers and their impact on clinical outcome following treatment with ipilimumab in advanced melanoma patients. EXPERIMENTAL DESIGN Changes in blood counts and the frequency of circulating immune cell populations analyzed by flow cytometry were investigated in 82 patients to compare baseline values with different time-points after starting ipilimumab. Endpoints were overall survival (OS) and best clinical response. Statistical calculations were done by Wilcoxon-matched pairs tests, Fisher exact test, Kaplan-Meier analysis, and Cox regression analysis. RESULTS Increases in absolute lymphocyte counts (ALC) 2 to 8 weeks (P = 0.003) and in percentages of CD4+ and CD8+ T cells 8 to 14 weeks (P = 0.001 and P = 0.02) after the first dose of ipilimumab were correlated with improved survival. These associations did not meet significance criteria, when conservatively adjusted for multiple testing, but were additionally correlated with clinical responses (all P < 0.05). However, validation is required. Increases in all three factors were observed in 36% of patients, who had a favorable outcome and survival probabilities of 93.3% and 63.8% at 12 and 24 months, respectively. A partial or complete response was observed in 71% of these patients compared with only 8% in patients with decreases in ≥1 of the 3 factors, respectively. Changes of regulatory T cells or myeloid-derived suppressor cells were not associated with OS. CONCLUSIONS Increases of ALC observed 2 to 8 weeks after initiation of ipilimumab and delayed increases in CD4+ and CD8+ T cells reflect changes associated with positive outcome. These changes represent surrogate marker candidates and warrant further validation. Clin Cancer Res; 22(19); 4848-58. ©2016 AACR.
Collapse
Affiliation(s)
- Alexander Martens
- Department of Dermatology, University Medical Center, Tübingen, Germany. Department of Internal Medicine II, University Medical Center, Tübingen, Germany
| | - Kilian Wistuba-Hamprecht
- Department of Dermatology, University Medical Center, Tübingen, Germany. Department of Internal Medicine II, University Medical Center, Tübingen, Germany
| | - Jianda Yuan
- Memorial Sloan Kettering Cancer Center, New York, New York
| | - Michael A Postow
- Memorial Sloan Kettering Cancer Center, New York, New York. Weill Cornell Medical College, New York, New York
| | - Phillip Wong
- Memorial Sloan Kettering Cancer Center, New York, New York
| | | | | | - Amir Khammari
- Department of Oncodermatology, INSERM Research Unit 892, University Hospital, Nantes, France
| | - Bastian Schilling
- Department of Dermatology, University Hospital, West German Cancer Center, University Duisburg-Essen, Essen, Germany. German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Antje Sucker
- Department of Dermatology, University Hospital, West German Cancer Center, University Duisburg-Essen, Essen, Germany. German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Dirk Schadendorf
- Department of Dermatology, University Hospital, West German Cancer Center, University Duisburg-Essen, Essen, Germany. German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Peter Martus
- Departments of Clinical Epidemiology and Applied Biostatistics, University of Tübingen, Tübingen, Germany
| | - Brigitte Dreno
- Department of Oncodermatology, INSERM Research Unit 892, University Hospital, Nantes, France
| | | | - Jedd D Wolchok
- Memorial Sloan Kettering Cancer Center, New York, New York. Weill Cornell Medical College, New York, New York
| | - Graham Pawelec
- Department of Internal Medicine II, University Medical Center, Tübingen, Germany. School of Science and Technology, College of Arts and Science, Nottingham Trent University, Nottingham, United Kingdom
| | - Claus Garbe
- Department of Dermatology, University Medical Center, Tübingen, Germany
| | - Benjamin Weide
- Department of Dermatology, University Medical Center, Tübingen, Germany.
| |
Collapse
|
1707
|
Dotto GP, Rustgi AK. Squamous Cell Cancers: A Unified Perspective on Biology and Genetics. Cancer Cell 2016; 29:622-637. [PMID: 27165741 PMCID: PMC4870309 DOI: 10.1016/j.ccell.2016.04.004] [Citation(s) in RCA: 231] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Revised: 01/20/2016] [Accepted: 04/07/2016] [Indexed: 01/11/2023]
Abstract
Squamous cell carcinomas (SCCs) represent the most frequent human solid tumors and are a major cause of cancer mortality. These highly heterogeneous tumors arise from closely interconnected epithelial cell populations with intrinsic self-renewal potential inversely related to the stratified differentiation program. SCCs can also originate from simple or pseudo-stratified epithelia through activation of quiescent cells and/or a switch in cell-fate determination. Here, we focus on specific determinants implicated in the development of SCCs by recent large-scale genomic, genetic, and epigenetic studies, and complementary functional analysis. The evidence indicates that SCCs from various body sites, while clinically treated as separate entities, have common determinants, pointing to a unified perspective of the disease and potential new avenues for prevention and treatment.
Collapse
Affiliation(s)
- G Paolo Dotto
- Department of Biochemistry, University of Lausanne, Epalinges 1066, Switzerland; Cutaneous Biology Research Center, Massachusetts General Hospital, Charlestown, MA 02129, USA.
| | - Anil K Rustgi
- Division of Gastroenterology, Departments of Medicine and Genetics, Abramson Cancer Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA.
| |
Collapse
|
1708
|
Banzi M, De Blasio S, Lallas A, Longo C, Moscarella E, Alfano R, Argenziano G. Dabrafenib: a new opportunity for the treatment of BRAF V600-positive melanoma. Onco Targets Ther 2016; 9:2725-33. [PMID: 27226731 PMCID: PMC4866744 DOI: 10.2147/ott.s75104] [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] [Indexed: 01/10/2023] Open
Abstract
Prior to 2011, the 1-year survival rates for patients suffering from advanced or metastatic melanoma was as low as 33%, with a median overall survival of about 9 months. Several chemotherapeutic regimens have been applied, either as monochemotherapy or as polychemotherapy, overall not resulting in an improvement of progression-free or overall survival. Novel insights into the epidemiology and biology of melanoma allowed the development of newer therapies. The discovery of mutations in BRAF, a part of the mitogen-activated protein kinase, allowed the development of two BRAF inhibitors, vemurafenib and dabrafenib, which significantly improved the outcome of metastatic melanoma treatment. This article reviews the mechanism of action, efficacy, and safety profile of dabrafenib. An in-depth knowledge of this medication will encourage clinicians to select the appropriate therapeutic strategy for each patient, as well as to prevent or adequately manage side effects, optimizing, thus, the drug's applicability.
Collapse
Affiliation(s)
- Maria Banzi
- Department of Medical Oncology, Arcispedale Santa Maria Nuova IRCCS, Reggio Emilia, Italy
| | - Simona De Blasio
- Skin Cancer Unit, Arcispedale Santa Maria Nuova IRCCS, Reggio Emilia, Italy
| | - Aimilios Lallas
- First Department of Dermatology, Aristotle University, Thessaloniki, Greece
| | - Caterina Longo
- Skin Cancer Unit, Arcispedale Santa Maria Nuova IRCCS, Reggio Emilia, Italy
| | - Elvira Moscarella
- Skin Cancer Unit, Arcispedale Santa Maria Nuova IRCCS, Reggio Emilia, Italy
| | - Roberto Alfano
- Department of Anesthesiology, Surgery and Emergency, Second University of Naples, Naples, Italy
| | | |
Collapse
|
1709
|
Guennoun A, Sidahmed H, Maccalli C, Seliger B, Marincola FM, Bedognetti D. Harnessing the immune system for the treatment of melanoma: current status and future prospects. Expert Rev Clin Immunol 2016; 12:879-93. [PMID: 27070898 DOI: 10.1080/1744666x.2016.1176529] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
When malignant melanoma is diagnosed early, surgical resection is the intervention of choice and is often curative, but many patients present with unresectable disease at later stages. Due to its complex etiology paired with well-documented chemoresistance and high metastatic potential, patients with advanced melanoma had a poor prognosis, and the treatment of this disease remained unsatisfactory for many years. Recently, targeted therapy, immune checkpoint inhibition, or combinatory approaches have revolutionized the therapeutic options of melanoma allowing considerable improvement in disease control and survival. In this review we will summarize these novel therapeutic strategies with particular focus on combinatory immunotherapies and further discuss recent data derived from immunogenomic studies and potential options to improve the therapeutic efficacy of immune modulatory approaches.
Collapse
Affiliation(s)
- Andrea Guennoun
- a Division of Translational Medicine , Research Branch, Sidra Medical and Research Center , Doha , Qatar
| | - Heba Sidahmed
- a Division of Translational Medicine , Research Branch, Sidra Medical and Research Center , Doha , Qatar
| | - Cristina Maccalli
- b Tumor Biology, Immunology and Therapy Section, Division of Translational Medicine , Research Branch, Sidra Medical and Research Center , Doha , Qatar
| | - Barbara Seliger
- c Institute of Medical Immunology , Martin Luther University Halle-Wittenberg , Halle , Germany
| | - Francesco M Marincola
- d Office of the Chief Research Officer (CRO) , Research Branch, Sidra Medical and Research Center , Doha , Qatar
| | - Davide Bedognetti
- b Tumor Biology, Immunology and Therapy Section, Division of Translational Medicine , Research Branch, Sidra Medical and Research Center , Doha , Qatar
| |
Collapse
|
1710
|
Le Tourneau C, Dreno B, Kirova Y, Grob JJ, Jouary T, Dutriaux C, Thomas L, Lebbé C, Mortier L, Saiag P, Avril MF, Maubec E, Joly P, Bey P, Cosset JM, Sun JS, Asselain B, Devun F, Marty ME, Dutreix M. First-in-human phase I study of the DNA-repair inhibitor DT01 in combination with radiotherapy in patients with skin metastases from melanoma. Br J Cancer 2016; 114:1199-205. [PMID: 27140316 PMCID: PMC4891504 DOI: 10.1038/bjc.2016.120] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Revised: 03/11/2016] [Accepted: 04/08/2016] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND DT01 is a DNA-repair inhibitor preventing recruitment of DNA-repair enzymes at damage sites. Safety, pharmacokinetics and preliminary efficacy through intratumoural and peritumoural injections of DT01 were evaluated in combination with radiotherapy in a first-in-human phase I trial in patients with unresectable skin metastases from melanoma. METHODS Twenty-three patients were included and received radiotherapy (30 Gy in 10 sessions) on all selected tumour lesions, comprising of two lesions injected with DT01 three times a week during the 2 weeks of radiotherapy. DT01 dose levels of 16, 32, 48, 64 and 96 mg were used, in a 3+3 dose escalation design, with an expansion cohort at 96 mg. RESULTS The median follow-up was 180 days. All patients were evaluable for safety and pharmacokinetics. No dose-limiting toxicity was observed and the maximum-tolerated dose was not reached. Most frequent adverse events were reversible grades 1 and 2 injection site reactions. Pharmacokinetic analyses demonstrated a systemic passage of DT01. Twenty-one patients were evaluable for efficacy on 76 lesions. Objective response was observed in 45 lesions (59%), including 23 complete responses (30%). CONCLUSIONS Intratumoural and peritumoural DT01 in combination with radiotherapy is safe and pharmacokinetic analyses suggest a systemic passage of DT01.
Collapse
Affiliation(s)
- C Le Tourneau
- Department of Medical Oncology, Institut Curie, Paris & Saint-Cloud 75005, France.,EA7285, Versailles-Saint-Quentin-en-Yvelines University, Versailles 78000, France
| | - B Dreno
- CHU de Nantes-Hôtel Dieu, Nantes 44093, France
| | - Y Kirova
- Radiotherapy Department, Institut Curie, Paris 75005, France
| | - J J Grob
- La Timone Hospital-APHM, Aix-Marseille University, Marseille 13385, France
| | - T Jouary
- Dermatology department, Saint-André Hospital, CHU de Bordeaux, Bordeaux 33000, France
| | - C Dutriaux
- Dermatology department, Saint-André Hospital, CHU de Bordeaux, Bordeaux 33000, France
| | - L Thomas
- Lyon Sud Hospital Center, Lyon 1 University, Pierre Benite 69495, France
| | - C Lebbé
- Saint-Louis Hospital, APHP, Paris 75010, France
| | - L Mortier
- Dermatology department, CHRU of Lille, Lille 59037, France
| | - P Saiag
- Ambroise Paré Hospital, Boulogne Billancourt 92104, France
| | - M F Avril
- Cochin hospital, APHP, Paris 75014, France
| | - E Maubec
- Bichat Hospital, Paris 75877, France
| | - P Joly
- CHU Rouen, Charles-Nicolle, Rouen 76000, France
| | - P Bey
- Institut Curie, Paris 75005, France
| | - J M Cosset
- Radiotherapy Department, Institut Curie, Paris 75005, France
| | - J S Sun
- DNA Therapeutics, Evry 91058, France
| | - B Asselain
- Department of Biostatistics, Institut Curie, Paris 75005, France
| | - F Devun
- DNA Therapeutics, Evry 91058, France.,Institut Curie, Orsay 91405, France
| | - M E Marty
- Saint-Louis Hospital, APHP, Paris 75010, France
| | - M Dutreix
- Institut Curie, Orsay 91405, France.,CNRS-UMR3347, INSERM-U1021, Paris-Sud University, Orsay 91405, France
| |
Collapse
|
1711
|
A Nexus Consisting of Beta-Catenin and Stat3 Attenuates BRAF Inhibitor Efficacy and Mediates Acquired Resistance to Vemurafenib. EBioMedicine 2016; 8:132-149. [PMID: 27428425 PMCID: PMC4919613 DOI: 10.1016/j.ebiom.2016.04.037] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2016] [Revised: 04/13/2016] [Accepted: 04/28/2016] [Indexed: 02/06/2023] Open
Abstract
Acquired resistance to second generation BRAF inhibitors (BRAFis), like vemurafenib is limiting the benefits of long term targeted therapy for patients with malignant melanomas that harbor BRAF V600 mutations. Since many resistance mechanisms have been described, most of them causing a hyperactivation of the MAPK- or PI3K/AKT signaling pathways, one potential strategy to overcome BRAFi resistance in melanoma cells would be to target important common signaling nodes. Known factors that cause secondary resistance include the overexpression of receptor tyrosine kinases (RTKs), alternative splicing of BRAF or the occurrence of novel mutations in MEK1 or NRAS. In this study we show that β-catenin is stabilized and translocated to the nucleus in approximately half of the melanomas that were analyzed and which developed secondary resistance towards BRAFi. We further demonstrate that β-catenin is involved in the mediation of resistance towards vemurafenib in vitro and in vivo. Unexpectedly, β-catenin acts mainly independent of the TCF/LEF dependent canonical Wnt-signaling pathway in resistance development, which partly explains previous contradictory results about the role of β-catenin in melanoma progression and therapy resistance. We further demonstrate that β-catenin interacts with Stat3 after chronic vemurafenib treatment and both together cooperate in the acquisition and maintenance of resistance towards BRAFi.
Collapse
|
1712
|
Targeted therapy in BRAF-mutated lung adenocarcinoma. Lancet Oncol 2016; 17:550-1. [DOI: 10.1016/s1470-2045(16)00117-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2016] [Accepted: 02/17/2016] [Indexed: 11/24/2022]
|
1713
|
Kourie HR, Gharios J, Kattan J. Allied therapies against BRAF-mutated advanced colon cancer: the right plans to win the battle. COLORECTAL CANCER 2016. [DOI: 10.2217/crc-2016-0003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Hampig Raphael Kourie
- Oncology Department, Jules Bordet Institute, Brussels, Belgium
- Oncology Department, Faculty of Medicine, Saint Joseph University, Beirut, Lebanon
| | - Joseph Gharios
- Oncology Department, Faculty of Medicine, Saint Joseph University, Beirut, Lebanon
| | - Joseph Kattan
- Oncology Department, Faculty of Medicine, Saint Joseph University, Beirut, Lebanon
| |
Collapse
|
1714
|
Kim RS, Goossens N, Hoshida Y. Use of big data in drug development for precision medicine. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2016; 1:245-253. [PMID: 27430024 DOI: 10.1080/23808993.2016.1174062] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Drug development has been a costly and lengthy process with an extremely low success rate and lack of consideration of individual diversity in drug response and toxicity. Over the past decade, an alternative "big data" approach has been expanding at an unprecedented pace based on the development of electronic databases of chemical substances, disease gene/protein targets, functional readouts, and clinical information covering inter-individual genetic variations and toxicities. This paradigm shift has enabled systematic, high-throughput, and accelerated identification of novel drugs or repurposed indications of existing drugs for pathogenic molecular aberrations specifically present in each individual patient. The exploding interest from the information technology and direct-to-consumer genetic testing industries has been further facilitating the use of big data to achieve personalized Precision Medicine. Here we overview currently available resources and discuss future prospects.
Collapse
Affiliation(s)
- Rosa S Kim
- Division of Liver Diseases, Department of Medicine, Liver Cancer Program, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Nicolas Goossens
- Division of Liver Diseases, Department of Medicine, Liver Cancer Program, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, USA; Division of Gastroenterology and Hepatology, Geneva University Hospital, Geneva, Switzerland
| | - Yujin Hoshida
- Division of Liver Diseases, Department of Medicine, Liver Cancer Program, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, USA
| |
Collapse
|
1715
|
Abstract
The treatment of metastatic melanoma is rapidly changing. In 2002, the BRAF mutation was described in over 50% of melanomas and led to the first BRAF inhibitor, vemurafenib, being approved for clinical use in 2011. Clinical responses are often rapid but duration of response is limited due to the development of resistance. MEK is the next downstream target from BRAF in the MAP kinase pathway. Trametinib was the first MEK inhibitor to be approved for clinical use in 2013. Preclinical studies demonstrated a delay in resistance and a reduction in cutaneous toxicity by combined BRAF and MEK inhibition. Here, we review the rationale for clinical development of trametinib and give an update on recent clinical trials of trametinib alone and in combination with braf inhibition in melanoma.
Collapse
Affiliation(s)
- Neha Chopra
- Mount Vernon Hospital, Medical Oncology, Rickmansworth Road, Northwood, Middlesex, HA6 2RN, UK
| | | |
Collapse
|
1716
|
Detection of BRAF Mutations Using a Fully Automated Platform and Comparison with High Resolution Melting, Real-Time Allele Specific Amplification, Immunohistochemistry and Next Generation Sequencing Assays, for Patients with Metastatic Melanoma. PLoS One 2016; 11:e0153576. [PMID: 27111917 PMCID: PMC4844167 DOI: 10.1371/journal.pone.0153576] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Accepted: 03/31/2016] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Metastatic melanoma is a severe disease with one of the highest mortality rate in skin diseases. Overall survival has significantly improved with immunotherapy and targeted therapies. Kinase inhibitors targeting BRAF V600 showed promising results. BRAF genotyping is mandatory for the prescription of anti-BRAF therapies. METHODS Fifty-nine formalin-fixed paraffin-embedded melanoma samples were assessed using High-Resolution-Melting (HRM) PCR, Real-time allele-specific amplification (RT-ASA) PCR, Next generation sequencing (NGS), immunohistochemistry (IHC) and the fully-automated molecular diagnostics platform IdyllaTM. Sensitivity, specificity, positive predictive value and negative predictive value were calculated using NGS as the reference standard to compare the different assays. RESULTS BRAF mutations were found in 28(47.5%), 29(49.2%), 31(52.5%), 29(49.2%) and 27(45.8%) samples with HRM, RT-ASA, NGS, IdyllaTM and IHC respectively. Twenty-six (81.2%) samples were found bearing a c.1799T>A (p.Val600Glu) mutation, three (9.4%) with a c.1798_1799delinsAA (p.Val600Lys) mutation and one with c.1789_1790delinsTC (p.Leu597Ser) mutation. Two samples were found bearing complex mutations. CONCLUSIONS HRM appears the less sensitive assay for the detection of BRAF V600 mutations. The RT-ASA, IdyllaTM and IHC assays are suitable for routine molecular diagnostics aiming at the prescription of anti-BRAF therapies. IdyllaTM assay is fully-automated and requires less than 2 minutes for samples preparation and is the fastest of the tested assays.
Collapse
|
1717
|
Reinwald M, Boch T, Hofmann WK, Buchheidt D. Risk of Infectious Complications in Hemato-Oncological Patients Treated with Kinase Inhibitors. Biomark Insights 2016; 10:55-68. [PMID: 27127405 PMCID: PMC4841329 DOI: 10.4137/bmi.s22430] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Revised: 09/13/2015] [Accepted: 09/15/2015] [Indexed: 12/30/2022] Open
Abstract
Infectious complications are a major cause of morbidity and mortality in patients with hemato-oncological diseases. Although disease-related immunosuppression represents one factor, aggressive treatment regimens, such as chemotherapy, stem cell transplantation, or antibody treatment, account for a large proportion of infectious side effects. With the advent of targeted therapies affecting specific kinases in malignant diseases, the outcome of patients has further improved. Nonetheless, dependent on the specific pathway targeted or off-target activity of the kinase inhibitor, therapy-associated infectious complications may occur. We review the most common and approved kinase inhibitors targeting a variety of hemato-oncological malignancies for their immunosuppressive potential and evaluate their risk of infectious side effects based on preclinical evidence and clinical data in order to raise awareness of the potential risks involved.
Collapse
Affiliation(s)
- Mark Reinwald
- Department of Hematology and Oncology, Mannheim University Hospital, University of Heidelberg, Mannheim, Germany
| | - Tobias Boch
- Department of Hematology and Oncology, Mannheim University Hospital, University of Heidelberg, Mannheim, Germany
| | - Wolf-Karsten Hofmann
- Department of Hematology and Oncology, Mannheim University Hospital, University of Heidelberg, Mannheim, Germany
| | - Dieter Buchheidt
- Department of Hematology and Oncology, Mannheim University Hospital, University of Heidelberg, Mannheim, Germany
| |
Collapse
|
1718
|
Dillman RO. Long-Term Progression-Free and Overall Survival in Two Melanoma Patients Treated with Patient-Specific Therapeutic Vaccine Eltrapuldencel-T After Resection of a Solitary Liver Metastasis. Cancer Biother Radiopharm 2016; 31:71-4. [PMID: 27093340 DOI: 10.1089/cbr.2016.2003] [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: 11/12/2022] Open
Abstract
Hepatic metastases from melanoma are usually associated with recurrence and short survival, even in patients with a solitary metastasis. Two patients, one with melanoma of unknown primary and one with ocular melanoma, underwent resection of a solitary liver metastasis followed by treatment with eltrapuldencel-T, a patient-specific therapeutic vaccine consisting of autologous dendritic cells loaded with antigens from irradiated melanoma cells obtained from an autologous tumor cell line. Following surgical resection, the ocular melanoma patient remained progression free for more than 4.5 years and was known to be alive more than 8.5 years later, while the other patient, who previously had experienced lung and small bowel metastases, has remained disease free and is alive more than 12 years later. These two cases illustrate how immunotherapies designed to induce immune responses to tumor-associated antigens (TAA), as opposed to releasing previously existing responses to TAA that have been suppressed, may also enhance long-term disease control and survival.
Collapse
|
1719
|
Schreuer M, Meersseman G, Van Den Herrewegen S, Jansen Y, Chevolet I, Bott A, Wilgenhof S, Seremet T, Jacobs B, Buyl R, Maertens G, Neyns B. Quantitative assessment of BRAF V600 mutant circulating cell-free tumor DNA as a tool for therapeutic monitoring in metastatic melanoma patients treated with BRAF/MEK inhibitors. J Transl Med 2016; 14:95. [PMID: 27095081 PMCID: PMC4837559 DOI: 10.1186/s12967-016-0852-6] [Citation(s) in RCA: 112] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 04/06/2016] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND BRAF V600 mutant circulating cell-free tumor DNA (BRAF V600mut ctDNA) could serve as a specific biomarker in patients with BRAF V600 mutant melanoma. We analyzed the value of BRAF V600mut ctDNA from plasma as a monitoring tool for advanced melanoma patients treated with BRAF/MEK inhibitors. METHODS Allele-specific quantitative PCR analysis for BRAF V600 E/E2/D/K/R/M mutations was performed on DNA extracted from plasma of patients with known BRAF V600 mutant melanoma who were treated with dabrafenib and trametinib. RESULTS 245 plasma samples from 36 patients were analyzed. In 16 patients the first plasma sample was obtained before the first dosing of dabrafenib/trametinib. At baseline, BRAF V600mut ctDNA was detected in 75 % of patients (n = 12/16). BRAF V600mut ctDNA decreased rapidly upon initiation of targeted therapy (p < 0.001) and became undetectable in 60 % of patients (n = 7/12) after 6 weeks of treatment. During treatment, disease progression (PD) was diagnosed in 27 of 36 patients. An increase of the BRAF V600mut ctDNA copy number and fraction, identified PD with a sensitivity of 70 % (n = 19/27) and a specificity of 100 %. An increase in the BRAF V600mut ctDNA fraction was detected prior to clinical PD in 44 % of cases (n = 12/27) and simultaneously with PD in 26 % of patients (n = 7/27). CONCLUSIONS Quantitative analysis of BRAF V600mut ctDNA in plasma has unique features as a monitoring tool during treatment with BRAF/MEK inhibitors. Its potential as an early predictor of acquired resistance deserves further evaluation.
Collapse
Affiliation(s)
- Max Schreuer
- />Department of Medical Oncology, Universitair Ziekenhuis Brussel (UZ Brussel), Vrije Universiteit Brussel (VUB), Laarbeeklaan 101, 1090 Brussels, Belgium
| | - Geert Meersseman
- />Biocartis, Generaal De Wittelaan 11 B3, 2800 Mechelen, Belgium
| | | | - Yanina Jansen
- />Department of Medical Oncology, Universitair Ziekenhuis Brussel (UZ Brussel), Vrije Universiteit Brussel (VUB), Laarbeeklaan 101, 1090 Brussels, Belgium
| | - Ines Chevolet
- />Department of Dermatology, Universitair ziekenhuis Gent (UZ Gent), Universiteit Gent (UGent), De Pintelaan 185, 9000 Ghent, Belgium
| | - Ambre Bott
- />Department of Medical Oncology, Universitair Ziekenhuis Brussel (UZ Brussel), Vrije Universiteit Brussel (VUB), Laarbeeklaan 101, 1090 Brussels, Belgium
| | - Sofie Wilgenhof
- />Department of Medical Oncology, Universitair Ziekenhuis Brussel (UZ Brussel), Vrije Universiteit Brussel (VUB), Laarbeeklaan 101, 1090 Brussels, Belgium
| | - Teofila Seremet
- />Department of Medical Oncology, Universitair Ziekenhuis Brussel (UZ Brussel), Vrije Universiteit Brussel (VUB), Laarbeeklaan 101, 1090 Brussels, Belgium
| | - Bart Jacobs
- />Biocartis, Generaal De Wittelaan 11 B3, 2800 Mechelen, Belgium
| | - Ronald Buyl
- />Department of Biostatistics and Medical Informatics, Vrije Universiteit Brussel (VUB), Laarbeeklaan, 103, 1090 Brussels, Belgium
| | - Geert Maertens
- />Biocartis, Generaal De Wittelaan 11 B3, 2800 Mechelen, Belgium
| | - Bart Neyns
- />Department of Medical Oncology, Universitair Ziekenhuis Brussel (UZ Brussel), Vrije Universiteit Brussel (VUB), Laarbeeklaan 101, 1090 Brussels, Belgium
| |
Collapse
|
1720
|
Wick MR, Gru AA. Metastatic melanoma: Pathologic characterization, current treatment, and complications of therapy. Semin Diagn Pathol 2016; 33:204-18. [PMID: 27234321 DOI: 10.1053/j.semdp.2016.04.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Metastatic melanoma (MM) has the potential to involve virtually any anatomical site, and it also has a wide spectrum of histological appearances. General clinicopathologic data pertaining to MM are presented in this review, together with a discussion of its differential diagnosis and therapy. "Biological" agents used in the treatment of melanoma are considered, along with the pathological features of the complications that they may cause.
Collapse
Affiliation(s)
- Mark R Wick
- Division of Surgical Pathology & Cytopathology, University of Virginia Medical Center, Charlottesville, Virginia.
| | - Alejandro A Gru
- Division of Surgical Pathology & Cytopathology, University of Virginia Medical Center, Charlottesville, Virginia
| |
Collapse
|
1721
|
Galván-Banqueri M, Ubago-Pérez R, Molina-López T. The relative clinical efficacy of trametinib-dabrafenib and cobimetinib-vemurafenib in advanced melanoma: an indirect comparison. J Clin Pharm Ther 2016; 41:285-9. [PMID: 27079278 DOI: 10.1111/jcpt.12390] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 03/22/2016] [Indexed: 11/24/2022]
Abstract
WHAT IS KNOWN AND OBJECTIVE Melanoma causes the majority of skin cancer-related deaths. The outcome of melanoma depends on its stage at diagnosis. Currently, for patients with advanced melanoma, two MEK inhibitors (trametinib and cobimetinib) have been authorized by the European Medicines Agency. The main objective of this study was to compare the relative efficacy of trametinib-dabrafenib and cobimetinib-vemurafenib in patients with advanced melanoma through adjusted indirect treatment comparisons (ITCs). METHODS A search was made up to the 3rd of November 2015. Databases consulted were MEDLINE, the Cochrane Library and the Centre for Reviews and Dissemination. Randomized controlled trials (RCTs) which compared the efficacy of trametinib-dabrafenib or cobimetinib-vemurafenib versus a common treatment comparator, in which outcomes of overall survival, progression-free survival (PFS) and overall response rate (ORR) were considered. ITCs were carried out using the method proposed by Bucher et al. RESULTS AND DISCUSSION Two RCTs were included (one for each drugs combination). The results of the adjusted ITCs showed that there were no statistically significant differences between the two combinations in terms of PFS and ORR. WHAT IS NEW AND CONCLUSION The ITCs indicate no difference in efficacy between both treatments. However, there should be an independent, head-to-head trial of both combinations to confirm the results.
Collapse
Affiliation(s)
| | - R Ubago-Pérez
- Andalusian Agency for Health Technology Assessment, Seville, Spain
| | - T Molina-López
- Andalusian Agency for Health Technology Assessment, Seville, Spain
| |
Collapse
|
1722
|
Hölzel M, Tüting T. Inflammation-Induced Plasticity in Melanoma Therapy and Metastasis. Trends Immunol 2016; 37:364-374. [PMID: 27151281 DOI: 10.1016/j.it.2016.03.009] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 03/23/2016] [Accepted: 03/29/2016] [Indexed: 12/18/2022]
Abstract
Phenotype switching contributes to nongenomic heterogeneity in melanoma and other cancers. These dynamic and in part reversible phenotype changes impose diagnostic and therapeutic challenges. Understanding the reciprocal coevolution of melanoma and immune cell phenotypes during disease progression and in response to therapy is a prerequisite to improve current treatment strategies. Here we discuss how proinflammatory signals promote melanoma cell plasticity and govern interactions of melanoma and immune cells in the tumor microenvironment. We examine phenotypic plasticity and heterogeneity in different melanoma mouse models with respect to their utility for translational research and emphasize the interplay between melanoma cells and neutrophils as a critical driver of metastasis.
Collapse
Affiliation(s)
- Michael Hölzel
- Unit for RNA Biology, Department of Clinical Chemistry and Clinical Pharmacology, University of Bonn, 53105 Bonn, Germany.
| | - Thomas Tüting
- Department of Dermatology, University Hospital Magdeburg, 39120 Magdeburg, Germany.
| |
Collapse
|
1723
|
Tel J, Koornstra R, de Haas N, van Deutekom V, Westdorp H, Boudewijns S, van Erp N, Di Blasio S, Gerritsen W, Figdor CG, de Vries IJM, Hato SV. Preclinical exploration of combining plasmacytoid and myeloid dendritic cell vaccination with BRAF inhibition. J Transl Med 2016; 14:88. [PMID: 27075584 PMCID: PMC4831164 DOI: 10.1186/s12967-016-0844-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 03/30/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Melanoma is the most lethal type of skin cancer and its incidence is progressively increasing. The introductions of immunotherapy and targeted therapies have tremendously improved the treatment of melanoma. Selective inhibition of BRAF by vemurafenib results in objective clinical responses in around 50 % of patients suffering from BRAFV600 mutated melanoma. However, drug resistance often results in hampering long-term tumor control. Alternatively, immunotherapy by vaccination with natural dendritic cells (nDCs) demonstrated long-term tumor control in a proportion of patients. We postulate that the rapid tumor debulking by vemurafenib can synergize the long-term tumor control of nDC vaccination to result in an effective treatment modality in a large proportion of patients. Here, we investigated the feasibility of this combination by analyzing the effect of vemurafenib on the functionality of nDCs. METHODS Plasmacytoid DCs (pDCs) and myeloid DCs (mDCs) were isolated from PBMCs obtained from buffy coats from healthy volunteers or vemurafenib-treated melanoma patients. Maturation of pDCs, mDCs and immature monocyte-derived DCs was induced by R848 in the presence or absence of vemurafenib and analyzed by FACS. Cytokine production and T cell proliferation induced by mature DCs were analyzed. RESULTS Vemurafenib inhibited maturation and cytokine production of highly purified nDCs of healthy volunteers resulting in diminished allogeneic T cell proliferation. This deleterious effect of vemurafenib on nDC functionality was absent when total PBMCs were exposed to vemurafenib. In patients receiving vemurafenib, nDC functionality and T cell allostimulatory capacity were unaffected. CONCLUSION Although vemurafenib inhibited the functionality of purified nDC of healthy volunteers, this effect was not observed when nDCs were matured in the complete PBMC fraction. This might have been caused by increased vemurafenib uptake in absence of other cell types. In accordance, nDCs isolated from patients on active vemurafenib treatment showed no negative effects. In conclusion, our results pave the way for a combinatorial treatment strategy and, we propose that combining vemurafenib with nDC vaccination represent a powerful opportunity that deserves more investigation in the clinic.
Collapse
Affiliation(s)
- Jurjen Tel
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6500 HB, Nijmegen, The Netherlands
| | - Rutger Koornstra
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Nienke de Haas
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6500 HB, Nijmegen, The Netherlands
| | - Vincent van Deutekom
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6500 HB, Nijmegen, The Netherlands
| | - Harm Westdorp
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6500 HB, Nijmegen, The Netherlands.,Department of Medical Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Steve Boudewijns
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6500 HB, Nijmegen, The Netherlands.,Department of Medical Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Nielka van Erp
- Department of Pharmacy, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Stefania Di Blasio
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6500 HB, Nijmegen, The Netherlands
| | - Winald Gerritsen
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Carl G Figdor
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6500 HB, Nijmegen, The Netherlands
| | - I Jolanda M de Vries
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6500 HB, Nijmegen, The Netherlands.
| | - Stanleyson V Hato
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6500 HB, Nijmegen, The Netherlands
| |
Collapse
|
1724
|
Taylor AD, Micheel CM, Anderson IA, Levy MA, Lovly CM. The Path(way) Less Traveled: A Pathway-Oriented Approach to Providing Information about Precision Cancer Medicine on My Cancer Genome. Transl Oncol 2016; 9:163-165. [PMID: 27084433 PMCID: PMC4833964 DOI: 10.1016/j.tranon.2016.03.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 03/03/2016] [Indexed: 10/31/2022] Open
Abstract
This perspective describes the motivation, development, and implementation of pathway-based content for My Cancer Genome, an online precision medicine knowledge resource describing clinical implications of genetic alterations in cancer. As researchers uncover more about cancer pathogenesis, we are learning more not only about the specific genes and proteins involved but also about how those genes and proteins interact with others along cell signaling pathways. This knowledge has led researchers and clinicians to begin to think about cancer therapy using a pathway-based approach. To facilitate this approach, My Cancer Genome used a list of more than 800 cancer-related genes to identify 20 cancer-relevant pathways and then created content focused on demonstrating the therapeutic relevance of these pathways.
Collapse
Affiliation(s)
- Alexandria D. Taylor
- Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, 37232
| | - Christine M. Micheel
- Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, 37232,Department of Medicine, Division of Hematology and Oncology, Vanderbilt University School of Medicine, Nashville, TN, 37232
| | - Ingrid A. Anderson
- Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, 37232
| | - Mia A. Levy
- Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, 37232,Department of Medicine, Division of Hematology and Oncology, Vanderbilt University School of Medicine, Nashville, TN, 37232,Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, TN, 37232,Address all correspondence to: Mia A. Levy or Christine M. Lovly.
| | - Christine M. Lovly
- Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, 37232,Department of Medicine, Division of Hematology and Oncology, Vanderbilt University School of Medicine, Nashville, TN, 37232,Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN, 37232,Address all correspondence to: Mia A. Levy or Christine M. Lovly.
| |
Collapse
|
1725
|
Spain L, Julve M, Larkin J. Combination dabrafenib and trametinib in the management of advanced melanoma with BRAFV600 mutations. Expert Opin Pharmacother 2016; 17:1031-8. [PMID: 27027150 DOI: 10.1517/14656566.2016.1168805] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION In the 40-50% of advanced melanoma patients with tumors harboring BRAF V600E and V600 K mutations, BRAF inhibitors such as dabrafenib are a highly effective treatment. However, most patients develop resistance after several months on treatment. The addition of a MEK inhibitor, such as trametinib, to BRAF inhibition mitigates one key pathway of resistance, further increasing response rates and improving survival. AREAS COVERED This article summarizes the mechanism of action of the combination of dabrafenib and trametinib, its evolution through Phase I, II and III clinical trials and discusses its current use in the management of patients with advanced melanoma. EXPERT OPINION Combination therapy with dabrafenib and trametinib improves response rate, progression-free survival and overall survival when compared to dabrafenib or vemurafenib alone. The addition of trametinib to dabrafenib changes the adverse event profile, making hyperkeratosis and cutaneous squamous cell carcinomas less common but side effects such as fever and nausea more common. How dabrafenib/trametinib is best sequenced with other effective treatments such as immune checkpoint blockade remains uncertain.
Collapse
Affiliation(s)
- Lavinia Spain
- a Melanoma Unit, Royal Marsden Foundation NHS Trust , London , UK
| | - Maximilian Julve
- a Melanoma Unit, Royal Marsden Foundation NHS Trust , London , UK
| | - James Larkin
- a Melanoma Unit, Royal Marsden Foundation NHS Trust , London , UK
| |
Collapse
|
1726
|
Liu JS, Rao S. Long-term drug costs per life-month gained associated with first-line treatments for unresectable or metastatic melanoma. Exp Hematol Oncol 2016; 5:9. [PMID: 27069772 PMCID: PMC4827222 DOI: 10.1186/s40164-016-0039-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 03/30/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND For unresectable or metastatic melanoma, first-line ipilimumab has demonstrated long-term survival benefits over a 7-year period. First-line treatment with BRAF inhibitors has demonstrated efficacy in clinical trials with up to 3 years of follow-up. The long-term comparative efficacy and costs of ipilimumab and BRAF inhibitors are unknown. METHODS Patient-level data from 12 clinical studies for ipilimumab were used. Survival data were extracted from included clinical trials for BRAF inhibitors based on a systematic literature review. Different parametric survival models, including exponential, Gompertz, log-normal, and Weibull models, were used to fit reported overall survival (OS) data and to project long-term survival for BRAF inhibitors. Survival benefits were measured in terms of total life-months gained as calculated by the area under the curve of OS Kaplan-Meier curves for the observed ipilimumab data and projected BRAF inhibitor data. Total life-months gained and cumulative costs per life-month gained were compared between ipilimumab and BRAF inhibitors. RESULTS The systematic literature review identified six randomized-controlled trials of BRAF inhibitors for subsequent analyses. With 7-year follow-up, ipilimumab was associated with a total of 28.5 life-months gained. Based on the Weibull model, the extrapolated total life-months gained for BRAF inhibitors were 26.5 months for dabrafenib, 21.3 months for trametinib, 14.3 months for vemurafenib, and 24.6 months for dabrafenib + trametinib. In sensitivity analyses, extrapolated total life-months gained varied across the three other models, ranging from 13.7 to 36.8 months across therapies. Cumulative costs per life-month gained with ipilimumab decreased steadily over time, while the costs remained constant for BRAF inhibitors due to continuous dosing. By year 3, cumulative costs per life-month gained were the lowest with ipilimumab; by year 7, the costs were $4281 for ipilimumab, compared with $8920 for dabrafenib, $10,211 for trametinib, $11,002 for vemurafenib, and $19,132 for the dabrafenib + trametinib combination therapy. CONCLUSIONS Ipilimumab was associated with a better long-term cost-per-life month compared to BRAF agents. Long-term extrapolation of survival with BRAF agents was uncertain, and showed no evidence of prolonged survival compared to ipilimumab.
Collapse
Affiliation(s)
- Jun S. Liu
- Department of Statistics, Harvard University, Cambridge, MA USA
| | | |
Collapse
|
1727
|
Klempner SJ, Gershenhorn B, Tran P, Lee TK, Erlander MG, Gowen K, Schrock AB, Morosini D, Ross JS, Miller VA, Stephens PJ, Ou SHI, Ali SM. BRAFV600E Mutations in High-Grade Colorectal Neuroendocrine Tumors May Predict Responsiveness to BRAF-MEK Combination Therapy. Cancer Discov 2016; 6:594-600. [PMID: 27048246 DOI: 10.1158/2159-8290.cd-15-1192] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 04/01/2016] [Indexed: 12/24/2022]
Abstract
UNLABELLED Neuroendocrine tumors comprise a heterogeneous group of malignancies with a broad spectrum of clinical behavior. Poorly differentiated tumors follow an aggressive course with limited treatment options, and new approaches are needed. Oncogenic BRAF V600E (BRAF(V600E)) substitutions are observed primarily in melanoma, colon cancer, and non-small cell lung cancer, but have been identified in multiple tumor types. Here, we describe the first reported recurrent BRAF(V600E) mutations in advanced high-grade colorectal neuroendocrine tumors and identify a BRAF alteration frequency of 9% in 108 cases. Among these BRAF alterations, 80% were BRAF(V600E) Dramatic response to BRAF-MEK combination therapy occurred in two cases of metastatic high-grade rectal neuroendocrine carcinoma refractory to standard therapy. Urinary BRAF(V600E) circulating tumor DNA monitoring paralleled disease response. Our series represents the largest study of genomic profiling in colorectal neuroendocrine tumors and provides strong evidence that BRAF(V600E) is an oncogenic driver responsive to BRAF-MEK combination therapy in this molecular subset. SIGNIFICANCE BRAF(V600E) is an established oncogenic driver, but significant disparities in response exist among tumor types. Two patients with treatment-refractory high-grade colorectal neuroendocrine tumors harboring BRAF(V600E) exhibited rapid and durable response to combined BRAF-MEK inhibition, providing the first clinical evidence of efficacy in this aggressive tumor type. Cancer Discov; 6(6); 594-600. ©2016 AACR.This article is highlighted in the In This Issue feature, p. 561.
Collapse
Affiliation(s)
- Samuel J Klempner
- Division of Hematology-Oncology, Department of Medicine, Chao Family Comprehensive Cancer Center, University of California, Irvine, School of Medicine, Orange, California.
| | - Bruce Gershenhorn
- Cancer Treatment Centers of America, Midwestern Regional Medical Center, Zion, Illinois
| | - Phu Tran
- Division of Hematology-Oncology, Department of Medicine, Chao Family Comprehensive Cancer Center, University of California, Irvine, School of Medicine, Orange, California
| | - Thomas K Lee
- Department of Pathology, University of California, Irvine, Orange, California
| | | | - Kyle Gowen
- Foundation Medicine, Inc., Cambridge, Massachusetts
| | | | | | - Jeffrey S Ross
- Foundation Medicine, Inc., Cambridge, Massachusetts. Albany Medical College, Albany, New York
| | | | | | - Sai-Hong Ignatius Ou
- Division of Hematology-Oncology, Department of Medicine, Chao Family Comprehensive Cancer Center, University of California, Irvine, School of Medicine, Orange, California
| | - Siraj M Ali
- Foundation Medicine, Inc., Cambridge, Massachusetts
| |
Collapse
|
1728
|
Komatsubara KM, Manson DK, Carvajal RD. Selumetinib for the treatment of metastatic uveal melanoma: past and future perspectives. Future Oncol 2016; 12:1331-44. [PMID: 27044592 DOI: 10.2217/fon-2015-0075] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Uveal melanoma is a rare but aggressive subtype of melanoma. Nearly 50% of patients will develop metastatic disease despite primary enucleation or radiation therapy. There is currently no standard of care therapy for metastatic uveal melanoma, and no therapy that has been shown to prolong overall survival. Uveal melanoma is characterized by activation of signaling pathways including the MAPK pathway and the PI3K/AKT pathway, among others, via mutations in the G-α-proteins GNAQ and GNA11. MEK inhibition with selumetinib has been evaluated as a therapeutic strategy in metastatic uveal melanoma. This review will discuss preclinical and clinical studies evaluating selumetinib in metastatic uveal melanoma, as well as potential future perspectives on MEK inhibition in the management of metastatic uveal melanoma.
Collapse
Affiliation(s)
| | - Daniel K Manson
- Division of Hematology/Oncology, Columbia University Medical Center, New York, NY, USA
| | - Richard D Carvajal
- Division of Hematology/Oncology, Columbia University Medical Center, New York, NY, USA
| |
Collapse
|
1729
|
Abstract
Recent data have demonstrated improved survival with targeted and immune therapies in patients with advanced melanoma, leading to much excitement amongst the oncology community and the widespread use of these drugs in combination regimens. However, the place of these combination therapies in the treatment of advanced melanoma remains to be fully determined. In this perspectives article, we critically review the available data and outline the rationale for these combinations being adopted as the standard of care for patients with advanced melanoma in the future.
Collapse
Affiliation(s)
- Keiran S M Smalley
- The Department of Cutaneous Oncology, Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL, USA.
- The Department of Tumor Biology, Moffitt Cancer Center and Research Institute, Tampa, FL, USA.
| | - Zeynep Eroglu
- The Department of Cutaneous Oncology, Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL, USA
| | - Vernon K Sondak
- The Department of Cutaneous Oncology, Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL, USA
| |
Collapse
|
1730
|
Oncogene status as a diagnostic tool in ocular and cutaneous melanoma. Eur J Cancer 2016; 57:112-7. [DOI: 10.1016/j.ejca.2016.01.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 01/15/2016] [Indexed: 11/24/2022]
|
1731
|
Kaliks RA. An update on clinical oncology for the non-oncologist. EINSTEIN-SAO PAULO 2016; 14:294-9. [PMID: 27462901 PMCID: PMC4943365 DOI: 10.1590/s1679-45082016md3550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 11/13/2015] [Indexed: 11/22/2022] Open
Abstract
ABSTRACTRecent advances in the understanding of tumor driver mutations, signaling pathways that lead to tumor progression, and the better understanding of the interaction between tumor cells and the immune system are revolutionizing cancer treatment. The pace at which new treatments are approved and the prices at which they are set have made it even more difficult to offer these treatments in countries like Brazil. In this review we present for the non-oncologist these new treatments and compare their availability in Brazilian public health system and private health system with that of developed countries.RESUMOAvanços recentes na compreensão de mutações promotoras de desenvolvimento do câncer, sinalização que leva à progressão de tumores, e o avanço no entendimento da interação entre as células tumorais e o sistema imunológico estão revolucionando o tratamento do câncer. A velocidade com que novos tratamentos são aprovados e o alto custo das medicações dificultam a disponibilização de terapêuticas em países como o Brasil. Nesta revisão, apresentamos ao não oncologista esses novos tratamentos e comparamos sua disponibilidade nos sistemas público e privado de saúde no Brasil com os países desenvolvidos.
Collapse
|
1732
|
Hassel JC, Lee SB, Meiss F, Meier F, Dimitrakopoulou-Strauss A, Jäger D, Enk AH. Vemurafenib and ipilimumab: A promising combination? Results of a case series. Oncoimmunology 2016; 5:e1101207. [PMID: 27141385 PMCID: PMC4839308 DOI: 10.1080/2162402x.2015.1101207] [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] [Received: 08/10/2015] [Revised: 09/22/2015] [Accepted: 09/23/2015] [Indexed: 10/22/2022] Open
Abstract
The purpose of combining targeted agents and immunotherapy is to achieve a chance of long-term tumor control in highly advanced patients. Between April 2012 and December 2013, 10 patients with metastatic melanoma were treated with a combination treatment of vemurafenib and ipilimumab as an individual treatment decision after detailed information and giving written informed consent. All the patients had advanced symptomatic disease, seven with elevated serum lactate dehydrogenase (LDH) levels and six with brain metastases on MRI. After clinical improvement under vemurafenib monotherapy (median 11.5 weeks), four cycles of ipilimumab were administered additionally. Combination treatment was tolerated well, with only two patients developing ≥ grade 3 elevation of transaminases; this was asymptomatic and resolved on cessation of BRAF inhibitor treatment. Staging 12 weeks after initiation of ipilimumab revealed partial response for five patients, stable disease for two, and disease progression for three. Of the seven patients with disease control, we stopped vemurafenib for five, to determine whether ipilimumab treatment led to disease control. Two revealed progressive disease 2 mo later, and received vemurafenib again, but for three the disease was controlled for at least a year, and two are still in partial remission without any further treatment. Progression-free survival was a median of 8.0 mo (95% CI 4.8-11.2), overall survival (OS) was 13.0 mo (95% CI 5.0-21.0), and four patients are still alive. In conclusion, the combination of vemurafenib and ipilimumab was well tolerated and clinical outcome was promising. The combination of targeted and immunotherapies is currently addressed in clinical trials.
Collapse
Affiliation(s)
- Jessica C. Hassel
- Department of Dermatology and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
| | - Sophia B. Lee
- Department of Dermatology and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
| | - Frank Meiss
- Department of Dermatology, Medical Center, University of Freiburg, Freiburg, Germany
| | - Friedegund Meier
- Department of Dermatology, University Hospital Tübingen, Tübingen, Germany
| | | | - Dirk Jäger
- Department of Medical Oncology and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
| | - Alexander H. Enk
- Department of Dermatology and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
1733
|
Babacan NA. Will we have a new ending for Irvin Yalom's novel? A medical oncologist view. JOURNAL OF ONCOLOGICAL SCIENCES 2016. [DOI: 10.1016/j.jons.2016.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
1734
|
Sullivan R. Serum lactate dehydrogenase is a more useful biomarker of prognosis than serum S100B in patients with BRAF-mutant melanoma. Br J Dermatol 2016; 174:716-7. [DOI: 10.1111/bjd.14503] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- R.J. Sullivan
- Center for Melanoma; Massachusetts General Hospital Cancer Center; 55 Fruit Street Boston MA 02114 U.S.A
| |
Collapse
|
1735
|
Tang R, Xu X, Yang W, Yu W, Hou S, Xuan Y, Tang Z, Zhao S, Chen Y, Xiao X, Huang W, Guo W, Li M, Deng W. MED27 promotes melanoma growth by targeting AKT/MAPK and NF-κB/iNOS signaling pathways. Cancer Lett 2016; 373:77-87. [PMID: 26797421 DOI: 10.1016/j.canlet.2016.01.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 12/28/2015] [Accepted: 01/06/2016] [Indexed: 01/07/2023]
Abstract
The inhibitors of BRAF and MEK targeting MAPK signaling pathway provide a comparatively effective therapeutic strategy for melanoma caused by BRAF mutation. However, melanoma, especially metastatic melanoma, has become one of the most threatening malignancies. Thus, the identification of exact molecular mechanisms and the key components involved in such mechanisms is urgently needed in order to provide new therapeutic options for patients with melanoma. Here, we identified MED27 as a potential melanoma target and explored its role and the associated molecular mechanism involved in melanoma progression. MED27 was found to be highly expressed in melanoma cells and tumor tissues. Its silencing led to melanoma cell proliferation inhibition, cell cycle arrest and apoptosis induction accompanied by the inactivation of PI3K/AKT and MAPK/ERK signaling and the activation of Bax/Cyto-C/Caspase-dependent apoptotic pathway. In addition, silencing of MED27 led to the decrease of iNOS expression through inhibiting the activation of a serial of upstream key proteins of NF-κB signaling pathway and the translocation of p50/p65 from cytoplasm to nucleus. MED27 was also found to be able to interact with NF-κB and p300 and to be acetylated by p300. Furthermore, the results in a xenograft tumor model indicated that melanoma progression was effectively suppressed by MED27 knockdown accompanied by the down-regulation of p-AKT, p-ERK, p-MEK1/2, MMP-9, Bcl-2 and iNOS expressions in the tumor tissues. Taken together, our study not only demonstrated the new function of MED27 as an oncogenic protein and the associated molecular mechanisms involved in melanoma progression, but also provided a possibility for the development of MED27 as a new anticancer target in melanoma therapy.
Collapse
Affiliation(s)
- Ranran Tang
- Institute of Cancer Stem Cell & The Second Affiliated Hospital, Dalian Medical University, Dalian, China; Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Xiangdong Xu
- Department of Thyroid & Breast Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wenjing Yang
- Institute of Cancer Stem Cell & The Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Wendan Yu
- Institute of Cancer Stem Cell & The Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Shuai Hou
- Institute of Cancer Stem Cell & The Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Yang Xuan
- Institute of Cancer Stem Cell & The Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Zhipeng Tang
- Institute of Cancer Stem Cell & The Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Shilei Zhao
- Institute of Cancer Stem Cell & The Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Yiming Chen
- Institute of Cancer Stem Cell & The Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Xiangsheng Xiao
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Wenlin Huang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China; State Key Laboratory of Targeted Drug for Tumors of Guangdong Province, Guangzhou Double Bioproduct Inc., Guangzhou, China
| | - Wei Guo
- Institute of Cancer Stem Cell & The Second Affiliated Hospital, Dalian Medical University, Dalian, China.
| | - Man Li
- Institute of Cancer Stem Cell & The Second Affiliated Hospital, Dalian Medical University, Dalian, China.
| | - Wuguo Deng
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China; State Key Laboratory of Targeted Drug for Tumors of Guangdong Province, Guangzhou Double Bioproduct Inc., Guangzhou, China.
| |
Collapse
|
1736
|
Sloot S, Speijers M, Bastiaannet E, Hoekstra H. Is there a relation between type of primary melanoma treatment and the development of intralymphatic metastasis? A review of the literature. Cancer Treat Rev 2016; 45:120-8. [DOI: 10.1016/j.ctrv.2016.02.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 02/19/2016] [Accepted: 02/24/2016] [Indexed: 10/22/2022]
|
1737
|
Loh KP, Mondo E, Hansen EA, Sievert L, Fung C, Sahasrabudhe DM, Guancial E. Targeted Therapy Based on Tumor Genomic Analyses in Metastatic Urachal Carcinoma. Clin Genitourin Cancer 2016; 14:e449-52. [PMID: 27102403 DOI: 10.1016/j.clgc.2016.03.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 03/19/2016] [Indexed: 12/22/2022]
Affiliation(s)
- Kah Poh Loh
- Division of Hematology/Oncology, James P. Wilmot Cancer Institute, University of Rochester, Strong Memorial Hospital, Rochester, NY.
| | - Esther Mondo
- Division of Hematology/Oncology, James P. Wilmot Cancer Institute, University of Rochester, Strong Memorial Hospital, Rochester, NY
| | - Elizabeth A Hansen
- Department of Pharmacy, University of Rochester, Strong Memorial Hospital, Rochester, NY
| | - Lynn Sievert
- Division of Hematology/Oncology, James P. Wilmot Cancer Institute, University of Rochester, Strong Memorial Hospital, Rochester, NY
| | - Chunkit Fung
- Division of Hematology/Oncology, James P. Wilmot Cancer Institute, University of Rochester, Strong Memorial Hospital, Rochester, NY
| | - Deepak M Sahasrabudhe
- Division of Hematology/Oncology, James P. Wilmot Cancer Institute, University of Rochester, Strong Memorial Hospital, Rochester, NY
| | - Elizabeth Guancial
- Division of Hematology/Oncology, James P. Wilmot Cancer Institute, University of Rochester, Strong Memorial Hospital, Rochester, NY
| |
Collapse
|
1738
|
Spagnolo F, Picasso V, Spano L, Tanda E, Venzano C, Queirolo P. Update on Metastatic Uveal Melanoma: Progress and Challenges. BioDrugs 2016; 30:161-72. [DOI: 10.1007/s40259-016-0167-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
1739
|
Cost Effectiveness of Sequencing 34 Cancer-Associated Genes as an Aid for Treatment Selection in Patients with Metastatic Melanoma. Mol Diagn Ther 2016; 19:169-77. [PMID: 25926090 PMCID: PMC4469775 DOI: 10.1007/s40291-015-0140-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Objective To determine whether a next-generation sequencing (NGS) panel of 34 cancer-associated genes would cost-effectively aid in the treatment selection for patients with metastatic melanoma, compared with a single-site BRAF V600 mutation test. Methods A decision model was developed to estimate the costs and health outcomes of the two test strategies. The cost effectiveness of these two strategies was analyzed from a payer perspective over a 2-year time horizon with model parameters taken from the literature. Results In the base case, the gene sequencing panel strategy resulted in a cost of US$120,022 and 0.721 quality-adjusted life years (QALYs) per patient, whereas the single-site mutation test strategy resulted in a cost of US$128,965 and 0.704 QALYs. Thus, the gene sequencing panel strategy cost US$8943 less per patient and increased QALYs by 0.0174 per patient. Sensitivity analyses showed that, compared with the single-site mutation test strategy, the gene sequencing panel strategy had a 90.9 % chance of having reduced costs and increased QALYs, with the cost of the gene sequencing panel test having minimal effect on the incremental cost. Conclusion Compared with the single-site mutation test, the use of an NGS panel of 34 cancer-associated genes as an aid in selecting therapy for patients with metastatic melanoma reduced costs and increased QALYs. If the base-case results were applied to the 8900 patients diagnosed with metastatic melanoma in the USA each year, the gene sequencing panel strategy could result in an annual savings of US$79.6 million and a gain of 155 QALYs. Electronic supplementary material The online version of this article (doi:10.1007/s40291-015-0140-9) contains supplementary material, which is available to authorized users.
Collapse
|
1740
|
Keating GM. Cobimetinib Plus Vemurafenib: A Review in BRAF V600 Mutation-Positive Unresectable or Metastatic Melanoma. Drugs 2016; 76:605-15. [DOI: 10.1007/s40265-016-0562-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
1741
|
Abstract
Basic research in cancer biology, genetics and immunology has resulted in improved insights into mechanisms that drive tumor initiation and growth. This improved biologic understanding of the diseases we treat has led to unprecedented therapeutic breakthroughs across multiple tumor types. In this article, we discuss opportunities and challenges in contemporary cancer drug development, highlighting efficacy endpoints, clinical trial design and the thoughtful inclusion of the patient perspective. As the field re-examines old practices and explores new opportunities, we must continue to efficiently utilize the human and scientific resources at our disposal to foster the development and delivery of safe and effective therapies to cancer patients.
Collapse
Affiliation(s)
- Paul G Kluetz
- Office of Hematology and Oncology Products, FDA/CDER/OND, US Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, MD 20993, USA.
| | - Richard Pazdur
- Office of Hematology and Oncology Products, FDA/CDER/OND, US Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, MD 20993, USA
| |
Collapse
|
1742
|
Slimano F, Roessle C, Blanc C, De Maleissye MF, Bauler S. [Updates on prevention and treatment of melanoma: Pharmacist involvements and challenges]. ANNALES PHARMACEUTIQUES FRANÇAISES 2016; 74:335-49. [PMID: 26968263 DOI: 10.1016/j.pharma.2016.01.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 01/25/2016] [Accepted: 01/27/2016] [Indexed: 02/07/2023]
Abstract
Melanoma is a skin cancer that represents an actual public health problem. Its incidence is increasing every year. Environmental risk factors have been clearly identified. Early diagnosis of a suspicious skin lesion should be possible by any health professionals because the prognosis is correlated with the evolution of the disease and the presence of metastases. The advent of new therapies in metastatic forms with the development of immunotherapies and kinases inhibitors has significantly changed the management of this disease. New therapies are available in retail pharmacies and involve health professionals out of the hospital. This article is intended for community and hospital pharmacists and summarizes recommendations for primary and secondary prevention. It updates on new targeted therapies. It wants to give advices to the community pharmacists about the effective use of those treatments for melanoma.
Collapse
Affiliation(s)
- F Slimano
- Département de pharmacie clinique, Gustave-Roussy cancer campus, 114, rue Édouard-Vaillant, 94805 Villejuif, France; Laboratoire de pharmacologie et pharmacocinétique, UFR de pharmacie, université de Reims-Champagne-Ardenne, 51, rue Cognacq-Jay, 51100 Reims, France; Unité MEDyC, UMR CNRS/URCA, université de Reims-Champagne-Ardenne, 51, rue Cognacq-Jay, 51100 Reims, France.
| | - C Roessle
- Département de pharmacie clinique, Gustave-Roussy cancer campus, 114, rue Édouard-Vaillant, 94805 Villejuif, France
| | - C Blanc
- Service de pharmacie, hôpital Ambroise-Paré, 9, avenue Charles-de-Gaulle, 92104 Boulogne-Billancourt, France
| | - M-F De Maleissye
- Service de dermatologie, hôpital Ambroise-Paré, 9, avenue Charles-de-Gaulle, 92104 Boulogne-Billancourt, France
| | - S Bauler
- Service de pharmacie, hôpital Ambroise-Paré, 9, avenue Charles-de-Gaulle, 92104 Boulogne-Billancourt, France
| |
Collapse
|
1743
|
Hoos A. Development of immuno-oncology drugs - from CTLA4 to PD1 to the next generations. Nat Rev Drug Discov 2016; 15:235-47. [PMID: 26965203 DOI: 10.1038/nrd.2015.35] [Citation(s) in RCA: 460] [Impact Index Per Article: 51.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Since the regulatory approval of ipilimumab in 2011, the field of cancer immunotherapy has been experiencing a renaissance. This success is based on progress in both preclinical and clinical science, including the development of new methods of investigation. Immuno-oncology has become a sub-specialty within oncology owing to its unique science and its potential for substantial and long-term clinical benefit. Immunotherapy agents do not directly attack the tumour but instead mobilize the immune system - this can be achieved through various approaches that utilize adaptive or innate immunity. Therefore, immuno-oncology drug development encompasses a broad range of agents, including antibodies, peptides, proteins, small molecules, adjuvants, cytokines, oncolytic viruses, bi-specific molecules and cellular therapies. This Perspective summarizes the recent history of cancer immunotherapy, including the factors that led to its success, provides an overview of novel drug-development considerations, summarizes three generations of immunotherapies that have been developed since 2011 and, thus, illustrates the breadth of opportunities these new generations of immunotherapies represent.
Collapse
Affiliation(s)
- Axel Hoos
- Oncology Research and Development, GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, Pennsylvania 19426, USA
| |
Collapse
|
1744
|
Fedorenko IV, Abel EV, Koomen JM, Fang B, Wood ER, Chen YA, Fisher KJ, Iyengar S, Dahlman KB, Wargo JA, Flaherty KT, Sosman JA, Sondak VK, Messina JL, Gibney GT, Smalley KS. Fibronectin induction abrogates the BRAF inhibitor response of BRAF V600E/PTEN-null melanoma cells. Oncogene 2016; 35:1225-35. [PMID: 26073081 PMCID: PMC4679729 DOI: 10.1038/onc.2015.188] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Revised: 03/30/2015] [Accepted: 04/28/2015] [Indexed: 12/30/2022]
Abstract
The mechanisms by which some melanoma cells adapt to Serine/threonine-protein kinase B-Raf (BRAF) inhibitor therapy are incompletely understood. In the present study, we used mass spectrometry-based phosphoproteomics to determine how BRAF inhibition remodeled the signaling network of melanoma cell lines that were BRAF mutant and PTEN null. Short-term BRAF inhibition was associated with marked changes in fibronectin-based adhesion signaling that were PTEN dependent. These effects were recapitulated through BRAF siRNA knockdown and following treatment with chemotherapeutic drugs. Increased fibronectin expression was also observed in mouse xenograft models as well as specimens from melanoma patients undergoing BRAF inhibitor treatment. Analysis of a melanoma tissue microarray showed loss of PTEN expression to predict for a lower overall survival, with a trend for even lower survival being seen when loss of fibronectin was included in the analysis. Mechanistically, the induction of fibronectin limited the responses of these PTEN-null melanoma cell lines to vemurafenib, with enhanced cytotoxicity observed following the knockdown of either fibronectin or its receptor α5β1 integrin. This in turn abrogated the cytotoxic response to BRAF inhibition via increased AKT signaling, which prevented the induction of cell death by maintaining the expression of the pro-survival protein Mcl-1. The protection conveyed by the induction of FN expression could be overcome through combined treatment with a BRAF and PI3K inhibitor.
Collapse
Affiliation(s)
- Inna V. Fedorenko
- The Department of Molecular Oncology, The Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL, 33612
| | - Ethan V. Abel
- Department of Cancer Biology, Thomas Jefferson University, 233 South 10th Street, Philadelphia, PA, 19107, USA
| | - John M. Koomen
- The Department of Molecular Oncology, The Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL, 33612
| | - Bin Fang
- Department of Proteomics, The Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL, 33612
| | - Elizabeth R. Wood
- The Department of Molecular Oncology, The Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL, 33612
- Department of Proteomics, The Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL, 33612
| | - Y. Ann Chen
- Department of Biostatistics and Bioinformatics, The Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL, 33612
| | - Kate J. Fisher
- Department of Biostatistics and Bioinformatics, The Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL, 33612
| | - Sanjana Iyengar
- The Department of Cutaneous Oncology, The Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL, 33612
| | - Kimberly B. Dahlman
- Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 777 Research Building, Nashville, TN 37232
| | | | | | - Jeffrey A. Sosman
- Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, 2220 Pierce Avenue, 777 Research Building, Nashville, TN 37232
| | - Vernon K. Sondak
- The Department of Cutaneous Oncology, The Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL, 33612
| | - Jane L. Messina
- The Department of Cutaneous Oncology, The Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL, 33612
| | - Geoffrey T. Gibney
- The Department of Cutaneous Oncology, The Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL, 33612
| | - Keiran S.M. Smalley
- The Department of Molecular Oncology, The Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL, 33612
- The Department of Cutaneous Oncology, The Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL, 33612
| |
Collapse
|
1745
|
Cocorocchio E, Gandini S, Alfieri S, Battaglia A, Pennacchioli E, Tosti G, Spadola G, Barberis M, Leo MD, Riviello C, Pala L, Intelisano A, Martinoli C, Ferrucci PF. Dabrafenib in metastatic melanoma: a monocentric 'real life' experience. Ecancermedicalscience 2016; 10:624. [PMID: 26981153 PMCID: PMC4778691 DOI: 10.3332/ecancer.2016.624] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Indexed: 01/19/2023] Open
Abstract
Dabrafenib is a potent BRAF-kinase inhibitor. Its activity was evaluated on 40 consecutive metastatic melanoma patients (pts) harboring the V600BRAF mutations. Dabrafenib was administered orally at the dosage of 150 mg b.i.d. daily. ORR was 82%, with 7% CR, 62% PR, 13% SD and 18% PD. The median PFS and OS were seven and 17 months, respectively (median follow-up: 8.5 months). Increased risk of progression was found in pts with elevated LDH, ECOG PS >1 and more than two metastatic sites. Grade 3–4 adverse events were recorded in 4 pts. In this retrospective analysis, Dabrafenib confirmed its role as the standard clinical option in metastatic melanoma pts.
Collapse
Affiliation(s)
- E Cocorocchio
- Medical Oncology of Melanoma and Sarcoma Division, Istituto Europeo di Oncologia, via Ripamonti 435, Milan 2014, Italy
| | - S Gandini
- Biostatistics Division, Istituto Europeo di Oncologia, via Ripamonti 435, Milan 2014, Italy
| | - S Alfieri
- Medical Oncology of Melanoma and Sarcoma Division, Istituto Europeo di Oncologia, via Ripamonti 435, Milan 2014, Italy
| | - A Battaglia
- Medical Oncology of Melanoma and Sarcoma Division, Istituto Europeo di Oncologia, via Ripamonti 435, Milan 2014, Italy
| | - E Pennacchioli
- Sarcoma Unit, Istituto Europeo di Oncologia, via Ripamonti 435, Milan 2014, Italy
| | - G Tosti
- Dermatoncological Surgery Division, Istituto Europeo di Oncologia, via Ripamonti 435, Milan 2014, Italy
| | - G Spadola
- Dermatoncological Surgery Division, Istituto Europeo di Oncologia, via Ripamonti 435, Milan 2014, Italy
| | - M Barberis
- Pathology Division, Istituto Europeo di Oncologia, via Ripamonti 435, Milan 2014, Italy
| | - M Di Leo
- Medical Oncology of Melanoma and Sarcoma Division, Istituto Europeo di Oncologia, via Ripamonti 435, Milan 2014, Italy
| | - C Riviello
- Medical Oncology of Melanoma and Sarcoma Division, Istituto Europeo di Oncologia, via Ripamonti 435, Milan 2014, Italy
| | - L Pala
- Medical Oncology of Melanoma and Sarcoma Division, Istituto Europeo di Oncologia, via Ripamonti 435, Milan 2014, Italy
| | - A Intelisano
- Sarcoma Unit, Istituto Europeo di Oncologia, via Ripamonti 435, Milan 2014, Italy
| | - C Martinoli
- Medical Oncology of Melanoma and Sarcoma Division, Istituto Europeo di Oncologia, via Ripamonti 435, Milan 2014, Italy
| | - P F Ferrucci
- Medical Oncology of Melanoma and Sarcoma Division, Istituto Europeo di Oncologia, via Ripamonti 435, Milan 2014, Italy
| |
Collapse
|
1746
|
BRAF inhibition in hairy cell leukemia with low-dose vemurafenib. Blood 2016; 127:2847-55. [PMID: 26941398 DOI: 10.1182/blood-2015-11-680074] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Accepted: 02/13/2016] [Indexed: 11/20/2022] Open
Abstract
The activating mutation of the BRAF serine/threonine protein kinase (BRAF V600E) is the key driver mutation in hairy cell leukemia (HCL), suggesting opportunities for therapeutic targeting. We analyzed the course of 21 HCL patients treated with vemurafenib outside of trials with individual dosing regimens (240-1920 mg/d; median treatment duration, 90 days). Vemurafenib treatment improved blood counts in all patients, with platelets, neutrophils, and hemoglobin recovering within 28, 43, and 55 days (median), respectively. Complete remission was achieved in 40% (6/15 of evaluable patients) and median event-free survival was 17 months. Response rate and kinetics of response were independent of vemurafenib dosing. Retreatment with vemurafenib led to similar response patterns (n = 6). Pharmacodynamic analysis of BRAF V600E downstream targets showed that vemurafenib (480 mg/d) completely abrogated extracellular signal-regulated kinase phosphorylation of hairy cells in vivo. Typical side effects also occurred at low dosing regimens. We observed the development of acute myeloid lymphoma (AML) subtype M6 in 1 patient, and the course suggested disease acceleration triggered by vemurafenib. The phosphatidylinositol 3-kinase hotspot mutation (E545K) was identified in the AML clone, providing a potential novel mechanism for paradoxical BRAF activation. These data provide proof of dependence of HCL on active BRAF signaling. We provide evidence that antitumor and side effects are observed with 480 mg vemurafenib, suggesting that dosing regimens in BRAF-driven cancers could warrant reassessment in trials with implications for cost of cancer care.
Collapse
|
1747
|
Somasundaram R, Herlyn M, Wagner SN. The role of tumor microenvironment in melanoma therapy resistance. Melanoma Manag 2016; 3:23-32. [PMID: 30190870 PMCID: PMC6094607 DOI: 10.2217/mmt.15.37] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 10/06/2015] [Indexed: 12/16/2022] Open
Abstract
Melanoma patients develop resistance to both chemotherapy and targeted-therapy drugs. Promising preclinical and clinical results with immune checkpoint inhibitors using antibodies directed against cytotoxic T-lymphocyte-associated protein 4 and programmed cell death protein 1 have re-energized the field of immune-based therapies in melanoma. However, similar to chemotherapy or targeted therapies, immune checkpoint blockade responds in only subsets of melanoma patients. A number of factors, including gene mutations, altered cell-signaling pathways and tumor heterogeneity can contribute to therapy resistance. Recent studies have highlighted the role of inflammatory tumor microenvironment on therapy resistance of cancer cells. Cancer cells either alone or in conjunction with the tumor stroma can contribute to an inflammatory microenvironment. Multimodal approaches of targeting the tumor microenvironment, in addition to malignant cells, may be necessary for better therapy responses.
Collapse
Affiliation(s)
| | - Meenhard Herlyn
- The Wistar Institute, 3601 Spruce St, Philadelphia, PA 19104, USA
| | - Stephan N Wagner
- Division of Immunology, Allergy & Infectious Diseases (DIAID), Department of Dermatology, Medical University of Vienna, 1090 Wien, Austria
| |
Collapse
|
1748
|
Barbaric J, Sekerija M, Agius D, Coza D, Dimitrova N, Demetriou A, Safaei Diba C, Eser S, Gavric Z, Primic-Zakelj M, Zivkovic S, Zvolsky M, Bray F, Coebergh JW, Znaor A. Disparities in melanoma incidence and mortality in South-Eastern Europe: Increasing incidence and divergent mortality patterns. Is progress around the corner? Eur J Cancer 2016; 55:47-55. [PMID: 26773419 DOI: 10.1016/j.ejca.2015.11.019] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 10/23/2015] [Accepted: 11/18/2015] [Indexed: 11/27/2022]
Abstract
INTRODUCTION Most countries in South-Eastern Europe (SEE) have lower incidence, but higher mortality rates of malignant melanoma (MM) of the skin compared to North-Western Europe (NWE). We explored trends in MM incidence and mortality in SEE countries by sex and age and compared them with the trends in NWE. METHODS We obtained data on incident cases and deaths from MM (ICD-10 code C43) from 11 population-based cancer registries in Bosnia and Herzegovina, Bulgaria, Croatia, Cyprus, Czech Republic, Malta, Romania, Serbia, Slovakia, Slovenia and Turkey. We calculated age-specific rates for 25-49 ('young'), 50-69 ('middle aged') and 70+ years ('older') and estimated the average annual percent of change in incidence and mortality trends 2000-2010 according to age group and sex, using joinpoint regression analysis. FINDINGS The incidence rates of MM across the region were uniformly increasing. Significant increases in mortality rates were observed in middle aged men in Serbia and Bulgaria, middle aged women in Slovenia, older men in the Czech Republic, Serbia and Turkey, and older women in Slovenia and Serbia. INTERPRETATION While MM incidence rates were still increasing across SEE, mortality trends diverged and were less favourable than in NWE. Empowering cancer registration and improving the quality of incidence and mortality data will be essential for monitoring progress in MM control. In the context of prevention of melanoma, disparities in early detection appear to be widening the gap between SEE and NWE, while the provision of care to patients with advanced disease is likely to prove a challenge for regional healthcare budgets.
Collapse
Affiliation(s)
- Jelena Barbaric
- Agency for Quality and Accreditation in Health Care and Social Welfare, Department for Development, Research and Health Technology Assessment, Planinska 13, 10000 Zagreb, Croatia.
| | - Mario Sekerija
- Croatian National Cancer Registry, Croatian Institute of Public Health, Rockefellerova 7, 10000 Zagreb, Croatia.
| | - Dominic Agius
- Malta National Cancer Registry, Department for Health Information and Research, 95, G'Mangia Hill, Pieta, PTA 1313, Malta.
| | - Daniela Coza
- Regional Cancer Registry of Cluj, Oncological Institute 'Ion Chiricuta', Republicii Street 34-36, 400015 Cluj-Napoca, Romania.
| | - Nadya Dimitrova
- Bulgarian National Cancer Registry, National Oncology Hospital, 6, Plovdivsko Pole Street, 1756 Sofia, Bulgaria.
| | - Anna Demetriou
- Cyprus Cancer Registry-Health Monitoring Unit, Ministry of Health, 1 Prodromou Street & 17 Chilonos Street, 1448 Lefkosia, Cyprus.
| | - Chakameh Safaei Diba
- National Cancer Registry of Slovakia, National Health Information Center, Lazaretska 26, 81109 Bratislava, Slovakia.
| | - Sultan Eser
- Hacettepe University, Public Health Institute, Sıhhıye Campus, Sıhhıye, 06100 Ankara, Turkey; Izmir Cancer Registry, Izmir Public Health Directorate, Zubeyde Hanim Caddesi No:100, 35067 Karsiyaka, Izmir, Turkey.
| | - Zivana Gavric
- Cancer Registry of Republic of Srpska, The Public Health Institute Banja Luka, Jovan Ducic 1, 78 000 Banja Luka, Bosnia and Herzegovina.
| | - Maja Primic-Zakelj
- Cancer Registry of Republic of Slovenia, Oncology Institute of Oncology Ljubljana, Zaloska 2, 1000 Ljubljana, Slovenia.
| | - Snezana Zivkovic
- Cancer Registry of Central Serbia, Institute of Public Health of Serbia, Dr Subotica 5, 11000 Belgrade, Serbia.
| | - Miroslav Zvolsky
- Czech National Cancer Registry, Institute of Health Information and Statistics of the Czech Republic, Palackého nám. 4, PO Box 60, 128 01 Praha 2, Czech Republic.
| | - Freddie Bray
- Section of Cancer Surveillance, International Agency for Research on Cancer, 150 Cours Albert Thomas, 69372 CEDEX 08 Lyon, France.
| | - Jan Willem Coebergh
- Department of Public Health, Erasmus University Medical Center, PO Box 2040 3000 CA Rotterdam, The Netherlands.
| | - Ariana Znaor
- Section of Cancer Surveillance, International Agency for Research on Cancer, 150 Cours Albert Thomas, 69372 CEDEX 08 Lyon, France.
| |
Collapse
|
1749
|
Abstract
Twenty-five years ago, on the 75th anniversary of the Johns Hopkins Bloomberg School of Public Health, I noted that epidemiologic research was moving away from the traditional approaches used to investigate "epidemics" and their close relationship with preventive medicine. Twenty-five years later, the role of epidemiology as an important contribution to human population research, preventive medicine, and public health is under substantial pressure because of the emphasis on "big data," phenomenology, and personalized medical therapies. Epidemiology is the study of epidemics. The primary role of epidemiology is to identify the epidemics and parameters of interest of host, agent, and environment and to generate and test hypotheses in search of causal pathways. Almost all diseases have a specific distribution in relation to time, place, and person and specific "causes" with high effect sizes. Epidemiology then uses such information to develop interventions and test (through clinical trials and natural experiments) their efficacy and effectiveness. Epidemiology is dependent on new technologies to evaluate improved measurements of host (genomics), epigenetics, identification of agents (metabolomics, proteomics), new technology to evaluate both physical and social environment, and modern methods of data collection. Epidemiology does poorly in studying anything other than epidemics and collections of numerators and denominators without specific hypotheses even with improved statistical methodologies.
Collapse
|
1750
|
Abstract
Immune checkpoint therapy has started a revolution in the field of oncology. The concept that the immune system plays a critical role in antitumor responses, which has been around for decades, has finally been proven and firmly established with elegant preclinical studies and dramatic clinical responses in patients as a result of antibodies that block inhibitory T-cell pathways. However, the clinical responses being achieved are only in a subset of patients, and more work is needed to provide a better understanding of the mechanisms that elicit tumor rejection, which will enable identification of appropriate biomarkers, reveal new targets, provide data to guide combination studies, and eventually dictate a platform that allows more patients to derive clinical benefit, including cures, with immune checkpoint therapy.
Collapse
Affiliation(s)
- Padmanee Sharma
- From the Departments of Immunology and Genitourinary Medical Oncology, M. D. Anderson Cancer Center, Houston, TX.P.S.'s work was supported by the SU2C-CRI Dream Team Cancer Immunotherapy Grant, PCF Challenge Grant in Immunology, NCI/NIH 1-R01 CA1633793-01, and Cancer Prevention Research in Texas (grant RP120108)
| |
Collapse
|