1751
|
Jenkins RW, Sullivan RJ. NRAS mutant melanoma: an overview for the clinician for melanoma management. Melanoma Manag 2016; 3:47-59. [PMID: 30190872 PMCID: PMC6097550 DOI: 10.2217/mmt.15.40] [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: 08/10/2015] [Accepted: 11/06/2015] [Indexed: 12/19/2022] Open
Abstract
Melanoma is the deadliest form of skin cancer and the incidence continues to rise in the United States and worldwide. Activating mutations in RAS oncogenes are found in roughly a third of all human cancers. Mutations in NRAS occur in approximately a fifth of cutaneous melanomas and are associated with aggressive clinical behavior. Cells harboring oncogenic NRAS mutations exhibit activation of multiple signaling cascades, including PI3K/Akt, MEK-ERK and RAL, which collectively stimulate cancer growth. While strategies to target N-Ras itself have proven ineffective, targeting one or more N-Ras effector pathways has shown promise in preclinical models. Despite promising preclinical data, current therapies for NRAS mutant melanoma remain limited. Immune checkpoint inhibitors and targeted therapies for BRAF mutant melanoma are transforming the treatment of metastatic melanoma, but the ideal treatment for NRAS mutant melanoma remains unknown. Improved understanding of NRAS mutant melanoma and relevant N-Ras effector signaling modules will be essential to develop new treatment strategies.
Collapse
Affiliation(s)
| | - Ryan J Sullivan
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
| |
Collapse
|
1752
|
|
1753
|
Abstract
Skin cancers represent a major public health problem. Cutaneous melanoma, the incidence of which is constantly increasing, has the particularity of metastasising when it is detected too late. Its treatment at the metastatic stage has evolved significantly over recent years with the arrival of targeted therapies and immunotherapies which can increase patients' survival.
Collapse
Affiliation(s)
- Barouyr Baroudjian
- Unité d'oncodermatologie, service de dermatologie du Professeur Bagot, Hôpital Saint-Louis, 1 avenue Claude-Vellefaux, 75010, Paris, France.
| | - Cécile Pagès
- Unité d'oncodermatologie, service de dermatologie du Professeur Bagot, Hôpital Saint-Louis, 1 avenue Claude-Vellefaux, 75010, Paris, France
| | - Céleste Lebbé
- Unité d'oncodermatologie, service de dermatologie du Professeur Bagot, Hôpital Saint-Louis, 1 avenue Claude-Vellefaux, 75010, Paris, France
| |
Collapse
|
1754
|
Medina PJ, Adams VR. PD-1 Pathway Inhibitors: Immuno-Oncology Agents for Restoring Antitumor Immune Responses. Pharmacotherapy 2016; 36:317-34. [PMID: 26822752 PMCID: PMC5071694 DOI: 10.1002/phar.1714] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Immune checkpoint inhibitors are designed to restore a patient's own antitumor immune response that has been suppressed during tumor development. The first monoclonal antibodies against the immune checkpoint programmed death 1 (PD-1) receptor, nivolumab and pembrolizumab, are now approved for clinical use. Both agents are indicated for the treatment of advanced melanoma, as well as for the treatment of metastatic non-small cell lung cancer (NSCLC). Nivolumab is also approved for the treatment of advanced renal cell carcinoma. In patients with melanoma, these agents result in objective response rates of ~25-40%, with durable responses lasting more than 2 years in some cases. Results from phase III trials have shown improved survival with nivolumab versus standard-of-care chemotherapy in both patients with advanced melanoma and those with advanced NSCLC. In patients with advanced melanoma, both PD-1 inhibitors (nivolumab and pembrolizumab) have shown improved survival versus ipilimumab. PD-1 inhibitors are associated with adverse events that have immune etiologies, with grade greater than 3 adverse events typically reported in 16% or less of patients. However, most immune-mediated adverse events (including grade 3-4 adverse events) can be managed by using published management algorithms without permanent discontinuation of the agent. As nivolumab and pembrolizumab enter the clinic, and with more PD-1 pathway agents in development for a range of tumor types, this review aims to provide pharmacists with a basic understanding of the role of PD-1 in modulating the immune system and their use in the cancer treatment. The most recent clinical efficacy and safety data are discussed, highlighting the response characteristics distinctive to immune checkpoint inhibitors, along with pharmacokinetic and pharmacodynamic data and cost considerations.
Collapse
Affiliation(s)
- Patrick J. Medina
- Department of Pharmacy: Clinical and Administrative SciencesThe University of Oklahoma College of PharmacyOklahoma CityOklahoma
| | - Val R. Adams
- Pharmacy Practice and Science DepartmentUniversity of Kentucky College of PharmacyLexingtonKentucky
| |
Collapse
|
1755
|
Asmar R, Yang J, Carvajal RD. Clinical utility of nivolumab in the treatment of advanced melanoma. Ther Clin Risk Manag 2016; 12:313-25. [PMID: 27013881 PMCID: PMC4778791 DOI: 10.2147/tcrm.s78039] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Melanomas are highly immunogenic tumors that evade the immune system by exploiting innate checkpoint pathways, rendering effector T-cells anergic. The immunotherapeutic approach of checkpoint inhibition can restore and invigorate endogenous antitumor T-cell responses and has become an important treatment option for patients with advanced melanoma. The CTLA-4 inhibitor ipilimumab and the PD-1 inhibitors nivolumab and pembrolizumab have been shown to induce durable responses and improve overall survival in metastatic, refractory melanoma. Optimization and validation of pretreatment biomarkers to predict response to these agents is a crucial area of ongoing research. Combination immunotherapy has recently demonstrated superior response rates compared to monotherapy; further investigation is needed to refine combinatorial strategies.
Collapse
Affiliation(s)
- Ramsey Asmar
- Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Jessica Yang
- Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Richard D Carvajal
- Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY, USA
- Department of Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA
| |
Collapse
|
1756
|
Smith LK, Rao AD, McArthur GA. Targeting metabolic reprogramming as a potential therapeutic strategy in melanoma. Pharmacol Res 2016; 107:42-47. [PMID: 26924126 DOI: 10.1016/j.phrs.2016.02.009] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Revised: 02/08/2016] [Accepted: 02/09/2016] [Indexed: 12/28/2022]
Abstract
Metabolic reprogramming is a recognized hallmark of cancer. In order to support continued proliferation and growth, tumor cells must metabolically adapt to balance their bioenergetic and biosynthetic needs. To achieve this, cancer cells switch from mitochondrial oxidative phosphorylation to predominantly rely on glycolysis, a process known as the "Warburg effect". The BRAF oncogene has recently emerged as a critical regulator of this process in melanoma, bringing to the fore the importance of metabolic reprogramming in the pathogenesis and treatment of metastatic melanoma. In this review, we summarize our current understanding of oncogenic reprogramming of metabolism in BRAF and NRAS mutant melanoma, and highlight emerging evidence supporting a metabolic basis for MAPK pathway inhibitor resistance and metabolic vulnerabilities that may be exploited to overcome this.
Collapse
Affiliation(s)
- Lorey K Smith
- Molecular Oncology Laboratory, Oncogenic Signaling and Growth Control Program, Peter MacCallum Cancer Centre, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Australia
| | - Aparna D Rao
- Molecular Oncology Laboratory, Oncogenic Signaling and Growth Control Program, Peter MacCallum Cancer Centre, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Australia
| | - Grant A McArthur
- Molecular Oncology Laboratory, Oncogenic Signaling and Growth Control Program, Peter MacCallum Cancer Centre, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Australia; Translational Research Laboratory, Cancer Therapeutics Program, Peter MacCallum Cancer Centre, Australia; Department of Pathology, University of Melbourne, Australia; Department of Medicine, St. Vincent's Hospital, University of Melbourne, Australia.
| |
Collapse
|
1757
|
Waizenegger IC, Baum A, Steurer S, Stadtmüller H, Bader G, Schaaf O, Garin-Chesa P, Schlattl A, Schweifer N, Haslinger C, Colbatzky F, Mousa S, Kalkuhl A, Kraut N, Adolf GR. A Novel RAF Kinase Inhibitor with DFG-Out-Binding Mode: High Efficacy in BRAF-Mutant Tumor Xenograft Models in the Absence of Normal Tissue Hyperproliferation. Mol Cancer Ther 2016; 15:354-65. [PMID: 26916115 DOI: 10.1158/1535-7163.mct-15-0617] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 12/30/2015] [Indexed: 11/16/2022]
Abstract
BI 882370 is a highly potent and selective RAF inhibitor that binds to the DFG-out (inactive) conformation of the BRAF kinase. The compound inhibited proliferation of human BRAF-mutant melanoma cells with 100× higher potency (1-10 nmol/L) than vemurafenib, whereas wild-type cells were not affected at 1,000 nmol/L. BI 882370 administered orally was efficacious in multiple mouse models of BRAF-mutant melanomas and colorectal carcinomas, and at 25 mg/kg twice daily showed superior efficacy compared with vemurafenib, dabrafenib, or trametinib (dosed to provide exposures reached in patients). To model drug resistance, A375 melanoma-bearing mice were initially treated with vemurafenib; all tumors responded with regression, but the majority subsequently resumed growth. Trametinib did not show any efficacy in this progressing population. BI 882370 induced tumor regression; however, resistance developed within 3 weeks. BI 882370 in combination with trametinib resulted in more pronounced regressions, and resistance was not observed during 5 weeks of second-line therapy. Importantly, mice treated with BI 882370 did not show any body weight loss or clinical signs of intolerability, and no pathologic changes were observed in several major organs investigated, including skin. Furthermore, a pilot study in rats (up to 60 mg/kg daily for 2 weeks) indicated lack of toxicity in terms of clinical chemistry, hematology, pathology, and toxicogenomics. Our results indicate the feasibility of developing novel compounds that provide an improved therapeutic window compared with first-generation BRAF inhibitors, resulting in more pronounced and long-lasting pathway suppression and thus improved efficacy.
Collapse
Affiliation(s)
- Irene C Waizenegger
- Department of Pharmacology and Translational Research, Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria.
| | - Anke Baum
- Department of Pharmacology and Translational Research, Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
| | - Steffen Steurer
- Department of Medicinal Chemistry, Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
| | - Heinz Stadtmüller
- Department of Medicinal Chemistry, Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
| | - Gerd Bader
- Department of Medicinal Chemistry, Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
| | - Otmar Schaaf
- Department of Discovery ADME, Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
| | - Pilar Garin-Chesa
- Department of Pharmacology and Translational Research, Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
| | - Andreas Schlattl
- Department of Pharmacology and Translational Research, Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
| | - Norbert Schweifer
- Department of Pharmacology and Translational Research, Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
| | - Christian Haslinger
- Department of Pharmacology and Translational Research, Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
| | - Florian Colbatzky
- Department of Non-clinical Drug Safety, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Sien Mousa
- Department of Non-clinical Drug Safety, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Arno Kalkuhl
- Department of Non-clinical Drug Safety, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Norbert Kraut
- Department of Pharmacology and Translational Research, Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
| | - Günther R Adolf
- Department of Pharmacology and Translational Research, Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
| |
Collapse
|
1758
|
Goulielmaki M, Koustas E, Moysidou E, Vlassi M, Sasazuki T, Shirasawa S, Zografos G, Oikonomou E, Pintzas A. BRAF associated autophagy exploitation: BRAF and autophagy inhibitors synergise to efficiently overcome resistance of BRAF mutant colorectal cancer cells. Oncotarget 2016; 7:9188-9221. [PMID: 26802026 PMCID: PMC4891035 DOI: 10.18632/oncotarget.6942] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 01/02/2016] [Indexed: 02/05/2023] Open
Abstract
Autophagy is the basic catabolic mechanism that involves cell degradation of unnecessary or dysfunctional cellular components. Autophagy has a controversial role in cancer--both in protecting against tumor progression by isolation of damaged organelles, or by potentially contributing to cancer growth. The impact of autophagy in RAS induced transformation still remains to be further analyzed based on the differential effect of RAS isoforms and tumor cell context. In the present study, the effect of KRAS/BRAF/PIK3CA oncogenic pathways on the autophagic cell properties and on main components of the autophagic machinery like p62 (SQSTM1), Beclin-1 (BECN1) and MAP1LC3 (LC3) in colon cancer cells was investigated. This study provides evidence that BRAF oncogene induces the expression of key autophagic markers, like LC3 and BECN1 in colorectal tumor cells. Herein, PI3K/AKT/MTOR inhibitors induce autophagic tumor properties, whereas RAF/MEK/ERK signalling inhibitors reduce expression of autophagic markers. Based on the ineffectiveness of BRAFV600E inhibitors in BRAFV600E bearing colorectal tumors, the BRAF related autophagic properties in colorectal cancer cells are further exploited, by novel combinatorial anti-cancer protocols. Strong evidence is provided here that pre-treatment of autophagy inhibitor 3-MA followed by its combination with BRAFV600E targeting drug PLX4720 can synergistically sensitize resistant colorectal tumors. Notably, colorectal cancer cells are very sensitive to mono-treatments of another autophagy inhibitor, Bafilomycin A1. The findings of this study are expected to provide novel efficient protocols for treatment of otherwise resistant colorectal tumors bearing BRAFV600E, by exploiting the autophagic properties induced by BRAF oncogene.
Collapse
Affiliation(s)
- Maria Goulielmaki
- Laboratory of Signal Mediated Gene Expression, Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, Athens, Greece
| | - Evangelos Koustas
- Laboratory of Signal Mediated Gene Expression, Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, Athens, Greece
| | - Eirini Moysidou
- Laboratory of Signal Mediated Gene Expression, Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, Athens, Greece
| | - Margarita Vlassi
- Laboratory of Signal Mediated Gene Expression, Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, Athens, Greece
| | | | - Senji Shirasawa
- Department of Cell Biology, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - George Zografos
- 3rd Department of Surgery, General Hospital of Athens G. Gennimatas, Athens, Greece
| | - Eftychia Oikonomou
- Laboratory of Signal Mediated Gene Expression, Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, Athens, Greece
| | - Alexander Pintzas
- Laboratory of Signal Mediated Gene Expression, Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, Athens, Greece
| |
Collapse
|
1759
|
Abstract
Immune checkpoint-blocking antibodies that enhance the immune system's ability to fight cancer are becoming important components of treatment for patients with a variety of malignancies. Cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) was the first immune checkpoint to be clinically targeted, and ipilimumab, an inhibitor of CTLA-4, was approved by the U.S. Food and Drug Administration (FDA) for patients with advanced melanoma. The programmed cell death-1 (PD-1) receptor and one of its ligands, PD-L1, more recently have shown great promise as therapeutic targets in a variety of malignancies. Nivolumab and pembrolizumab recently have been FDA- approved for patients with melanoma and additional approvals within this therapeutic class are expected. The use of anti-CTLA-4 and anti-PD-1/PD-L1 antibodies is associated with side effects known as immune-related adverse events (irAEs). Immune-related adverse events affect the dermatologic, gastrointestinal, hepatic, endocrine, and other organ systems. Temporary immunosuppression with corticosteroids, tumor necrosis factor-alpha antagonists, mycophenolate mofetil, or other agents can be effective treatment. This article describes the side-effect profile of the checkpoint-blocking antibodies that target CTLA-4 and PD-1/PD-L1 and provides suggestions on how to manage specific irAEs.
Collapse
Affiliation(s)
- Michael A Postow
- From the Memorial Sloan Kettering Cancer Center, New York, NY; Weill Cornell Medical College, New York, NY
| |
Collapse
|
1760
|
Cardiovascular Toxicity and Management of Targeted Cancer Therapy. Am J Med Sci 2016; 351:535-43. [PMID: 27140715 DOI: 10.1016/j.amjms.2016.02.023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 12/07/2015] [Indexed: 12/24/2022]
Abstract
The advent of effective oral, molecular-targeted drugs in oncology has changed many incurable malignancies such as chronic myeloid leukemia into chronic diseases similar to coronary artery disease and diabetes mellitus. Oral agents including monoclonal antibodies, kinase inhibitors and hormone receptor blockers offer patients with cancer incremental improvements in both overall survival and quality of life. As it is imperative to recognize and manage side effects of platelet inhibitors, beta blockers, statins, human immunodeficiency virus drugs and fluoroquinolones by all healthcare providers, the same holds true for these newer targeted therapies; patients may present to their generalist or other subspecialist with drug-related symptoms. Cardiovascular adverse events are among the most frequent, and potentially serious, health issues in outpatient clinics, and among the most frequent side effects of targeted chemotherapy. Data support improved patient outcomes and satisfaction when primary care and other providers are cognizant of chemotherapy side effects, allowing for earlier intervention and reduction in morbidity and healthcare costs. With the implementation of accountable care and pay for performance, improved communication between generalists and subspecialists is essential to deliver cost-effective patient care.
Collapse
|
1761
|
Ivashko IN, Kolesar JM. Pembrolizumab and nivolumab: PD-1 inhibitors for advanced melanoma. Am J Health Syst Pharm 2016; 73:193-201. [DOI: 10.2146/ajhp140768] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Affiliation(s)
| | - Jill M. Kolesar
- University of Wisconsin-Madison, WI, and Director, 3P Analytical Laboratory, University of Wisconsin Carbone Cancer Center, Madison, WI
| |
Collapse
|
1762
|
Tan J. Perianal melanoma with a BRAF gene mutation in a young Portuguese Roma native. BMJ Case Rep 2016; 2016:bcr-2015-212772. [PMID: 26880821 DOI: 10.1136/bcr-2015-212772] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
A case of a young man diagnosed with perianal nodular melanoma with a gene mutation, accompanied by regional and pulmonary metastases on initial presentation, and later on with hepatic and bone involvement, is presented. The patient underwent wide local excision but was unresponsive to dacarbazine. Targeted therapy with vemurafenib had shown clinical improvement for a 5-month duration until he showed signs of disease progression. Just after the shift of adjuvant therapy to ipilimumab, he was diagnosed with multiple cerebral metastases that eventually led to his demise 6 months after initiation of vemurafenib, having had a 12-month survival period from the time of initial melanoma diagnosis.
Collapse
|
1763
|
Kong BY, Carlino MS, Menzies AM. Biology and treatment of BRAF mutant metastatic melanoma. Melanoma Manag 2016; 3:33-45. [PMID: 30190871 DOI: 10.2217/mmt.15.38] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 10/12/2015] [Indexed: 12/31/2022] Open
Abstract
BRAF inhibitors were among the first systemic therapies to show clinical benefit in metastatic melanoma. Here, we review the spectrum of BRAF mutations in melanoma, their role in oncogenesis, clinicopathological associations and response to treatment. The differing biology and clinical features of V600E- and V600K-mutated melanoma are outlined. The molecular changes associated with BRAF fusion genes and their response to targeted therapies, as well as the role of immunotherapy in treatment sequencing with targeted therapies are discussed.
Collapse
Affiliation(s)
- Benjamin Y Kong
- Crown Princess Mary Cancer Care Centre, Westmead, Sydney, NSW 2145, Australia.,Crown Princess Mary Cancer Care Centre, Westmead, Sydney, NSW 2145, Australia
| | - Matteo S Carlino
- Crown Princess Mary Cancer Care Centre, Westmead, Sydney, NSW 2145, Australia.,Melanoma Institute Australia, North Sydney, NSW 2060, Australia.,Sydney Medical School, The University of Sydney, Sydney, NSW 2006, Australia.,Crown Princess Mary Cancer Care Centre, Westmead, Sydney, NSW 2145, Australia.,Melanoma Institute Australia, North Sydney, NSW 2060, Australia.,Sydney Medical School, The University of Sydney, Sydney, NSW 2006, Australia
| | - Alexander M Menzies
- Melanoma Institute Australia, North Sydney, NSW 2060, Australia.,Sydney Medical School, The University of Sydney, Sydney, NSW 2006, Australia.,Royal North Shore Hospital, Sydney, NSW 2065, Australia.,Melanoma Institute Australia, North Sydney, NSW 2060, Australia.,Sydney Medical School, The University of Sydney, Sydney, NSW 2006, Australia.,Royal North Shore Hospital, Sydney, NSW 2065, Australia
| |
Collapse
|
1764
|
Grünhagen DJ, Kroon HM, Verhoef C. Perfusion and infusion for melanoma in-transit metastases in the era of effective systemic therapy. Am Soc Clin Oncol Educ Book 2016:e528-34. [PMID: 25993219 DOI: 10.14694/edbook_am.2015.35.e528] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The management of melanoma in-transit metastases (IT-mets) is challenging. For many years, the absence of effective systemic therapy has prompted physicians to focus on regional therapies for melanoma confined to the limb. The introduction of isolated limb perfusion (ILP) and isolated limb infusion (ILI) has enabled effective delivery of cytotoxic drugs in an isolated circuit, so as to overcome systemic toxicity and maximize local response. Both techniques have evolved over years and both tumor necrosis factor (TNF)-alpha-based ILP and ILI have distinct indications. The development of new systemic treatment options for patients with melanoma in the past decade has shed a new light on melanoma therapy. The present manuscript focuses on the modern role of ILI and ILP in the treatment of patients with melanoma with in-transit metastases in the era of effective systemic therapy. The response and control rates of ILI/ILP are still superior to rates achieved with systemic agents. The extent of disease in patients with stage III disease, however, warrants effective systemic treatment to prolong survival. There is great potential in combining rapid response therapy such as ILI/ILP with systemic agents for sustainable response. Trial results are eagerly awaited.
Collapse
Affiliation(s)
| | - Hidde M Kroon
- From the Erasmus MC Cancer Institute, Rotterdam, Netherlands
| | | |
Collapse
|
1765
|
Tarhini AA. Neoadjuvant therapy for melanoma: a promising therapeutic approach and an ideal platform in drug development. Am Soc Clin Oncol Educ Book 2016:e535-42. [PMID: 25993220 DOI: 10.14694/edbook_am.2015.35.e535] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Patients with locoregionally advanced but surgically operable melanoma continue to carry a high risk of relapse and death despite the best available standard management approaches. Neoadjuvant studies targeting this patient population tested chemotherapy with temozolomide and biochemotherapy (BCT), in which BCT demonstrated high tumor response rates but was eventually abandoned with the failure of BCT to deliver survival benefits in randomized trials of metastatic disease. Smaller neoadjuvant immunotherapy studies with interferon (IFN) alfa and ipilimumab have yielded promising clinical activity and important mechanistic insights and biomarker findings. Newer targeted and immunotherapeutic agents and combinations currently are being translated into the neoadjuvant setting at an accelerated pace and carry significant clinical promise. In drug development, the neoadjuvant approach allows access to blood and tumor tissue before and after initiation of systemic therapy, which allows for the conduct of novel mechanistic and biomarker studies in the circulation and the tumor microenvironment. Such studies may guide drug development and allow for the discovery of predictive biomarkers selected on the basis of their capacity to classify patients according to the degree of benefit from treatment or the risk for significant toxicity.
Collapse
Affiliation(s)
- Ahmad A Tarhini
- From the University of Pittsburgh School of Medicine and Cancer Institute, Pittsburgh, PA
| |
Collapse
|
1766
|
Cooper ZA, Reuben A, Spencer CN, Prieto PA, Austin-Breneman JL, Jiang H, Haymaker C, Gopalakrishnan V, Tetzlaff MT, Frederick DT, Sullivan RJ, Amaria RN, Patel SP, Hwu P, Woodman SE, Glitza IC, Diab A, Vence LM, Rodriguez-Canales J, Parra ER, Wistuba II, Coussens LM, Sharpe AH, Flaherty KT, Gershenwald JE, Chin L, Davies MA, Clise-Dwyer K, Allison JP, Sharma P, Wargo JA. Distinct clinical patterns and immune infiltrates are observed at time of progression on targeted therapy versus immune checkpoint blockade for melanoma. Oncoimmunology 2016; 5:e1136044. [PMID: 27141370 PMCID: PMC4839346 DOI: 10.1080/2162402x.2015.1136044] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Revised: 12/17/2015] [Accepted: 12/18/2015] [Indexed: 12/20/2022] Open
Abstract
We have made major advances in the treatment of melanoma through the use of targeted therapy and immune checkpoint blockade; however, clinicians are posed with therapeutic dilemmas regarding timing and sequence of therapy. There is a growing appreciation of the impact of antitumor immune responses to these therapies, and we performed studies to test the hypothesis that clinical patterns and immune infiltrates differ at progression on these treatments. We observed rapid clinical progression kinetics in patients on targeted therapy compared to immune checkpoint blockade. To gain insight into possible immune mechanisms behind these differences, we performed deep immune profiling in tumors of patients on therapy. We demonstrated low CD8+ T-cell infiltrate on targeted therapy and high CD8+ T-cell infiltrate on immune checkpoint blockade at clinical progression. These data have important implications, and suggest that antitumor immune responses should be assessed when considering therapeutic options for patients with melanoma.
Collapse
Affiliation(s)
- Zachary A Cooper
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA; Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Alexandre Reuben
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center , Houston, TX, USA
| | - Christine N Spencer
- Genomic Medicine, University of Texas MD Anderson Cancer Center , Houston, TX, USA
| | - Peter A Prieto
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center , Houston, TX, USA
| | - Jacob L Austin-Breneman
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center , Houston, TX, USA
| | - Hong Jiang
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center , Houston, TX, USA
| | - Cara Haymaker
- Melanoma Medical Oncology, University of Texas MD Anderson Cancer Center , Houston, TX, USA
| | | | - Michael T Tetzlaff
- Pathology, University of Texas MD Anderson Cancer Center , Houston, TX, USA
| | - Dennie T Frederick
- Division of Medical Oncology, Massachusetts General Hospital , Boston, MA, USA
| | - Ryan J Sullivan
- Division of Medical Oncology, Massachusetts General Hospital , Boston, MA, USA
| | - Rodabe N Amaria
- Melanoma Medical Oncology, University of Texas MD Anderson Cancer Center , Houston, TX, USA
| | - Sapna P Patel
- Melanoma Medical Oncology, University of Texas MD Anderson Cancer Center , Houston, TX, USA
| | - Patrick Hwu
- Melanoma Medical Oncology, University of Texas MD Anderson Cancer Center , Houston, TX, USA
| | - Scott E Woodman
- Melanoma Medical Oncology, University of Texas MD Anderson Cancer Center , Houston, TX, USA
| | - Isabella C Glitza
- Melanoma Medical Oncology, University of Texas MD Anderson Cancer Center , Houston, TX, USA
| | - Adi Diab
- Melanoma Medical Oncology, University of Texas MD Anderson Cancer Center , Houston, TX, USA
| | - Luis M Vence
- Immunology, University of Texas MD Anderson Cancer Center , Houston, TX, USA
| | - Jaime Rodriguez-Canales
- Translational Molecular Pathology, University of Texas MD Anderson Cancer Center , Houston, TX, USA
| | - Edwin R Parra
- Translational Molecular Pathology, University of Texas MD Anderson Cancer Center , Houston, TX, USA
| | - Ignacio I Wistuba
- Translational Molecular Pathology, University of Texas MD Anderson Cancer Center , Houston, TX, USA
| | - Lisa M Coussens
- Department of Cell, Developmental and Cancer Biology and Knight Cancer Institute, Oregon Health and Science University , Portland, OR, USA
| | - Arlene H Sharpe
- Division of Immunology, Department of Microbiology and Immunobiology, Harvard Medical School , Boston, MA, USA
| | - Keith T Flaherty
- Division of Medical Oncology, Massachusetts General Hospital , Boston, MA, USA
| | - Jeffrey E Gershenwald
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center , Houston, TX, USA
| | - Lynda Chin
- Genomic Medicine, University of Texas MD Anderson Cancer Center , Houston, TX, USA
| | - Michael A Davies
- Melanoma Medical Oncology, University of Texas MD Anderson Cancer Center , Houston, TX, USA
| | - Karen Clise-Dwyer
- Stem Cell Transplantation, University of Texas MD Anderson Cancer Center , Houston, TX, USA
| | - James P Allison
- Immunology, University of Texas MD Anderson Cancer Center , Houston, TX, USA
| | - Padmanee Sharma
- Immunology, University of Texas MD Anderson Cancer Center, Houston, TX, USA; Genitourinary Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jennifer A Wargo
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA; Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
1767
|
Atkins MB, Larkin J. Immunotherapy Combined or Sequenced With Targeted Therapy in the Treatment of Solid Tumors: Current Perspectives. ACTA ACUST UNITED AC 2016; 108:djv414. [DOI: 10.1093/jnci/djv414] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 12/09/2015] [Indexed: 11/13/2022]
Affiliation(s)
- Michael B. Atkins
- Affiliations of authors: Georgetown-Lombardi Comprehensive Cancer Center , Washington, DC (MBA); Royal Marsden Hospital , London , UK (JL)
| | - James Larkin
- Affiliations of authors: Georgetown-Lombardi Comprehensive Cancer Center , Washington, DC (MBA); Royal Marsden Hospital , London , UK (JL)
| |
Collapse
|
1768
|
Akabane H, Sullivan RJ. The Future of Molecular Analysis in Melanoma: Diagnostics to Direct Molecularly Targeted Therapy. Am J Clin Dermatol 2016; 17:1-10. [PMID: 26518880 DOI: 10.1007/s40257-015-0159-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Melanoma is a malignancy of pigment-producing cells that is driven by a variety of genetic mutations and aberrations. In most cases, this leads to upregulation of the mitogen-activated protein kinase (MAPK) pathway through activating mutations of upstream mediators of the pathway including BRAF and NRAS. With the advent of effective MAPK pathway inhibitors, including the US FDA-approved BRAF inhibitors vemurafenib and dabrafenib and MEK inhibitor trametinib, molecular analysis has become an integral part of the care of patients with metastatic melanoma. In this article, the key molecular targets and strategies to inhibit these targets therapeutically are presented, and the techniques of identifying these targets, in both tissue and blood, are discussed.
Collapse
Affiliation(s)
- Hugo Akabane
- Department of Medicine, Metrowest Medical Center, Framingham, MA, USA
| | - Ryan J Sullivan
- Center for Melanoma, Massachusetts General Hospital Cancer Center, 55 Fruit Street, Boston, MA, 02114, USA.
| |
Collapse
|
1769
|
Vaidhyanathan S, Wilken-Resman B, Ma DJ, Parrish KE, Mittapalli RK, Carlson BL, Sarkaria JN, Elmquist WF. Factors Influencing the Central Nervous System Distribution of a Novel Phosphoinositide 3-Kinase/Mammalian Target of Rapamycin Inhibitor GSK2126458: Implications for Overcoming Resistance with Combination Therapy for Melanoma Brain Metastases. J Pharmacol Exp Ther 2016; 356:251-9. [PMID: 26604245 PMCID: PMC4727156 DOI: 10.1124/jpet.115.229393] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 11/23/2015] [Indexed: 12/25/2022] Open
Abstract
Small molecule inhibitors targeting the mitogen-activated protein kinase pathway (Braf/mitogen-activated protein kinase kinase/extracellular signal-regulated kinase) have had success in extending survival for patients with metastatic melanoma. Unfortunately, resistance may occur via cross-activation of alternate signaling pathways. One approach to overcome resistance is to simultaneously target the phosphoinositide 3-kinase/mammalian target of rapamycin signaling pathway. Recent reports have shown that GSK2126458 [2,4-difluoro-N-(2-methoxy-5-(4-(pyridazin-4-yl)quinolin-6-yl)pyridin-3-yl) benzenesulfonamide], a dual phosphoinositide 3-kinase/mammalian target of rapamycin inhibitor, can overcome acquired resistance to Braf and mitogen-activated protein kinase kinase inhibitors in vitro. These resistance mechanisms may be especially important in melanoma brain metastases because of limited drug delivery across the blood-brain barrier. The purpose of this study was to investigate factors that influence the brain distribution of GSK2126458 and to examine the efficacy of GSK2126458 in a novel patient-derived melanoma xenograft (PDX) model. Both in vitro and in vivo studies indicate that GSK2126458 is a substrate for P-glycoprotein (P-gp) and breast cancer resistance protein (Bcrp), two dominant active efflux transporters in the blood-brain barrier. The steady-state brain distribution of GSK2126458 was 8-fold higher in the P-gp/Bcrp knockout mice compared with the wild type. We also observed that when simultaneously infused to steady state, GSK212658, dabrafenib, and trametinib, a rational combination to overcome mitogen-activated protein kinase inhibitor resistance, all had limited brain distribution. Coadministration of elacridar, a P-gp/Bcrp inhibitor, increased the brain distribution of GSK2126458 by approximately 7-fold in wild-type mice. In the PDX model, GSK2126458 showed efficacy in flank tumors but was ineffective in intracranial melanoma. These results show that P-gp and Bcrp are involved in limiting the brain distribution of GSK2126458 and provide a rationale for the lack of efficacy of GSK2126458 in the orthotopic PDX model.
Collapse
Affiliation(s)
- Shruthi Vaidhyanathan
- Department of Pharmaceutics, Brain Barriers Research Center, University of Minnesota, Minneapolis, Minnesota (S.V., B.W.-R., K.E.P., R.K.M., W.F.E.); and Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (D.J.M, B.L.C., J.N.S.)
| | - Brynna Wilken-Resman
- Department of Pharmaceutics, Brain Barriers Research Center, University of Minnesota, Minneapolis, Minnesota (S.V., B.W.-R., K.E.P., R.K.M., W.F.E.); and Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (D.J.M, B.L.C., J.N.S.)
| | - Daniel J Ma
- Department of Pharmaceutics, Brain Barriers Research Center, University of Minnesota, Minneapolis, Minnesota (S.V., B.W.-R., K.E.P., R.K.M., W.F.E.); and Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (D.J.M, B.L.C., J.N.S.)
| | - Karen E Parrish
- Department of Pharmaceutics, Brain Barriers Research Center, University of Minnesota, Minneapolis, Minnesota (S.V., B.W.-R., K.E.P., R.K.M., W.F.E.); and Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (D.J.M, B.L.C., J.N.S.)
| | - Rajendar K Mittapalli
- Department of Pharmaceutics, Brain Barriers Research Center, University of Minnesota, Minneapolis, Minnesota (S.V., B.W.-R., K.E.P., R.K.M., W.F.E.); and Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (D.J.M, B.L.C., J.N.S.)
| | - Brett L Carlson
- Department of Pharmaceutics, Brain Barriers Research Center, University of Minnesota, Minneapolis, Minnesota (S.V., B.W.-R., K.E.P., R.K.M., W.F.E.); and Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (D.J.M, B.L.C., J.N.S.)
| | - Jann N Sarkaria
- Department of Pharmaceutics, Brain Barriers Research Center, University of Minnesota, Minneapolis, Minnesota (S.V., B.W.-R., K.E.P., R.K.M., W.F.E.); and Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (D.J.M, B.L.C., J.N.S.)
| | - William F Elmquist
- Department of Pharmaceutics, Brain Barriers Research Center, University of Minnesota, Minneapolis, Minnesota (S.V., B.W.-R., K.E.P., R.K.M., W.F.E.); and Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota (D.J.M, B.L.C., J.N.S.)
| |
Collapse
|
1770
|
Komatsubara KM, Carvajal RD. Selumetinib for the treatment of melanoma. Expert Opin Orphan Drugs 2016. [DOI: 10.1517/21678707.2016.1133281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
1771
|
|
1772
|
Boussemart L, Girault I, Malka-Mahieu H, Mateus C, Routier E, Rubington M, Kamsu-Kom N, Thomas M, Tomasic G, Agoussi S, Breckler M, Laporte M, Lacroix L, Eggermont AM, Cavalcanti A, Grange F, Adam J, Vagner S, Robert C. Secondary Tumors Arising in Patients Undergoing BRAF Inhibitor Therapy Exhibit Increased BRAF–CRAF Heterodimerization. Cancer Res 2016; 76:1476-84. [DOI: 10.1158/0008-5472.can-15-2900-t] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 12/13/2015] [Indexed: 11/16/2022]
|
1773
|
Dacarbazine in combination with bevacizumab for the treatment of unresectable/metastatic melanoma: a phase II study. Melanoma Res 2016; 25:239-45. [PMID: 25746039 DOI: 10.1097/cmr.0000000000000146] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The combined treatment of dacarbazine with an antiangiogenic drug such as bevacizumab may potentiate the therapeutic effects of dacarbazine in metastatic melanoma (MM). Preliminary antitumour activity of dacarbazine plus bevacizumab is evaluated, together with the toxicity and safety profile, in MM patients. This prospective, open-label, phase II study included patients with previously untreated MM or unresectable melanoma. Patients received dacarbazine and bevacizumab until progressive disease or unacceptable toxicity. The primary efficacy variable was the overall response rate. The secondary efficacy parameters included duration of response, duration of stable disease, time to progression/progression-free survival, time to treatment failure and overall survival. The safety analysis included recordings of adverse events and exposure to study treatment. The intention-to-treat population included 37 patients (24 men and 13 women, mean age 54.2±13.1 years). Overall response rate was 18.9% (seven patients achieved a response) and clinical benefit was 48.6%. In patients who achieved a response, the median duration of response was 16.9 months and the median duration of stable disease was 12.5 months. The median time to progression/progression-free survival and time to treatment failure were 5.5 and 3.1 months, respectively. The median overall survival was 11.4 months. Almost all patients (94.6%) experienced at least one adverse event; however, no new area of toxicity of bevacizumab emerged. The dacarbazine/bevacizumab combination provides benefits compared with dacarbazine monotherapy in historical controls, with an acceptable safety profile. This combination appears to be a valid option in specific subgroups of patients, namely, those triple negative (BRAF, C-KIT and NRAS wild type) or with a BRAF mutation who have already received, or are not eligible for, immunomodulating or targeted agents.
Collapse
|
1774
|
Affiliation(s)
- Tito Fojo
- Columbia University Medical Center, New York, NY.
| |
Collapse
|
1775
|
Brys AK, Gowda R, Loriaux DB, Robertson GP, Mosca PJ. Nanotechnology-based strategies for combating toxicity and resistance in melanoma therapy. Biotechnol Adv 2016; 34:565-577. [PMID: 26826558 DOI: 10.1016/j.biotechadv.2016.01.004] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2015] [Revised: 11/15/2015] [Accepted: 01/16/2016] [Indexed: 12/17/2022]
Abstract
Drug toxicity and resistance remain formidable challenges in cancer treatment and represent an area of increasing attention in the case of melanoma. Nanotechnology represents a paradigm-shifting field with the potential to mitigate drug resistance while improving drug delivery and minimizing toxicity. Recent clinical and pre-clinical studies have demonstrated how a diverse array of nanoparticles may be harnessed to circumvent known mechanisms of drug resistance in melanoma to improve therapeutic efficacy. In this review, we discuss known mechanisms of resistance to various melanoma therapies and possible nanotechnology-based strategies that could be used to overcome these barriers and improve the pharmacologic arsenal available to combat advanced stage melanoma.
Collapse
Affiliation(s)
- Adam K Brys
- Department of Surgery, Division of Surgical Oncology, Duke University Medical Center, Durham, NC 27710, United States
| | - Raghavendra Gowda
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, United States
| | - Daniel B Loriaux
- Department of Surgery, Division of Surgical Oncology, Duke University Medical Center, Durham, NC 27710, United States
| | - Gavin P Robertson
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, United States
| | - Paul J Mosca
- Department of Surgery, Division of Surgical Oncology, Duke University Medical Center, Durham, NC 27710, United States.
| |
Collapse
|
1776
|
Long GV, Weber JS, Infante JR, Kim KB, Daud A, Gonzalez R, Sosman JA, Hamid O, Schuchter L, Cebon J, Kefford RF, Lawrence D, Kudchadkar R, Burris HA, Falchook GS, Algazi A, Lewis K, Puzanov I, Ibrahim N, Sun P, Cunningham E, Kline AS, Del Buono H, McDowell DO, Patel K, Flaherty KT. Overall Survival and Durable Responses in Patients With BRAF V600-Mutant Metastatic Melanoma Receiving Dabrafenib Combined With Trametinib. J Clin Oncol 2016; 34:871-8. [PMID: 26811525 DOI: 10.1200/jco.2015.62.9345] [Citation(s) in RCA: 231] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE To report the overall survival (OS) and clinical characteristics of BRAF inhibitor-naive long-term responders and survivors treated with dabrafenib plus trametinib in a phase I and II study of patients with BRAF V600 mutation-positive metastatic melanoma. METHODS BRAF inhibitor-naive patients treated with dabrafenib 150 mg twice daily plus trametinib 2 mg daily (the 150/2 group) from the non-randomly assigned (part B) and randomly assigned (part C) cohorts of the study were analyzed for progression-free and OS separately. Baseline characteristics and factors on treatment were analyzed for associations with durable responses and OS. RESULTS For BRAF inhibitor-naive patients in the 150/2 groups (n = 78), the progression-free survival at 1, 2, and 3 years was 44%, 22%, and 18%, respectively, for part B (n = 24) and 41%, 25%, and 21%, respectively, for part C (n = 54). Median OS was 27.4 months in part B and 25 months in part C. OS at 1, 2, and 3 years was 72%, 60%, and 47%, respectively, for part B and 80%, 51%, and 38%, respectively, for part C. Prolonged survival was associated with metastases in fewer than three organ sites and lower baseline lactate dehydrogenase. OS at 3 years was 62% in patients with normal baseline lactate dehydrogenase and 63% in patients with a complete response. CONCLUSION Dabrafenib plus trametinib results in a median OS of more than 2 years in BRAF inhibitor-naive patients with BRAF V600 mutation-positive metastatic melanoma, and approximately 20% were progression free at 3 years. Durable responses occurred in patients with good prognostic features at baseline, which may be predictive.
Collapse
Affiliation(s)
- Georgina V Long
- Georgina V. Long, Melanoma Institute Australia; The University of Sydney; Richard F. Kefford, Melanoma Institute Australia; The University of Sydney; Macquarie University, Sydney; Westmead Hospital, Westmead; Jonathan Cebon, Austin Health, Melbourne, Victoria, Australia; Jeffrey S. Weber and Ragini Kudchadkar, Moffitt Cancer Center, Tampa, FL; Jeffrey R. Infante and Howard A. Burris III, Sarah Cannon Research Institute/Tennessee Oncology; Kevin B. Kim, California Pacific Medical Center; Adil Daud, Alain Algazi, University of California, San Francisco, San Francisco; Omid Hamid, The Angeles Clinic and Research Institute, Los Angeles, CA; Rene Gonzalez, Karl Lewis, University of Colorado; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver, CO; Jeffrey A. Sosman, Igor Puzanov, Vanderbilt University Medical Center, Nashville, TN; Lynn Schuchter, University of Pennsylvania Abramson Cancer Center; Nageatte Ibrahim, Elizabeth Cunningham, Merck; Peng Sun, Amy S. Kline, Heather Del Buono, Diane Opatt McDowell, GlaxoSmithKline, Philadelphia, PA; Donald Lawrence and Kiran Patel, Incyte Corporation, Wilmington, DE; and Keith T. Flaherty, Massachusetts General Hospital Cancer Center, Boston, MA.
| | - Jeffrey S Weber
- Georgina V. Long, Melanoma Institute Australia; The University of Sydney; Richard F. Kefford, Melanoma Institute Australia; The University of Sydney; Macquarie University, Sydney; Westmead Hospital, Westmead; Jonathan Cebon, Austin Health, Melbourne, Victoria, Australia; Jeffrey S. Weber and Ragini Kudchadkar, Moffitt Cancer Center, Tampa, FL; Jeffrey R. Infante and Howard A. Burris III, Sarah Cannon Research Institute/Tennessee Oncology; Kevin B. Kim, California Pacific Medical Center; Adil Daud, Alain Algazi, University of California, San Francisco, San Francisco; Omid Hamid, The Angeles Clinic and Research Institute, Los Angeles, CA; Rene Gonzalez, Karl Lewis, University of Colorado; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver, CO; Jeffrey A. Sosman, Igor Puzanov, Vanderbilt University Medical Center, Nashville, TN; Lynn Schuchter, University of Pennsylvania Abramson Cancer Center; Nageatte Ibrahim, Elizabeth Cunningham, Merck; Peng Sun, Amy S. Kline, Heather Del Buono, Diane Opatt McDowell, GlaxoSmithKline, Philadelphia, PA; Donald Lawrence and Kiran Patel, Incyte Corporation, Wilmington, DE; and Keith T. Flaherty, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Jeffrey R Infante
- Georgina V. Long, Melanoma Institute Australia; The University of Sydney; Richard F. Kefford, Melanoma Institute Australia; The University of Sydney; Macquarie University, Sydney; Westmead Hospital, Westmead; Jonathan Cebon, Austin Health, Melbourne, Victoria, Australia; Jeffrey S. Weber and Ragini Kudchadkar, Moffitt Cancer Center, Tampa, FL; Jeffrey R. Infante and Howard A. Burris III, Sarah Cannon Research Institute/Tennessee Oncology; Kevin B. Kim, California Pacific Medical Center; Adil Daud, Alain Algazi, University of California, San Francisco, San Francisco; Omid Hamid, The Angeles Clinic and Research Institute, Los Angeles, CA; Rene Gonzalez, Karl Lewis, University of Colorado; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver, CO; Jeffrey A. Sosman, Igor Puzanov, Vanderbilt University Medical Center, Nashville, TN; Lynn Schuchter, University of Pennsylvania Abramson Cancer Center; Nageatte Ibrahim, Elizabeth Cunningham, Merck; Peng Sun, Amy S. Kline, Heather Del Buono, Diane Opatt McDowell, GlaxoSmithKline, Philadelphia, PA; Donald Lawrence and Kiran Patel, Incyte Corporation, Wilmington, DE; and Keith T. Flaherty, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Kevin B Kim
- Georgina V. Long, Melanoma Institute Australia; The University of Sydney; Richard F. Kefford, Melanoma Institute Australia; The University of Sydney; Macquarie University, Sydney; Westmead Hospital, Westmead; Jonathan Cebon, Austin Health, Melbourne, Victoria, Australia; Jeffrey S. Weber and Ragini Kudchadkar, Moffitt Cancer Center, Tampa, FL; Jeffrey R. Infante and Howard A. Burris III, Sarah Cannon Research Institute/Tennessee Oncology; Kevin B. Kim, California Pacific Medical Center; Adil Daud, Alain Algazi, University of California, San Francisco, San Francisco; Omid Hamid, The Angeles Clinic and Research Institute, Los Angeles, CA; Rene Gonzalez, Karl Lewis, University of Colorado; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver, CO; Jeffrey A. Sosman, Igor Puzanov, Vanderbilt University Medical Center, Nashville, TN; Lynn Schuchter, University of Pennsylvania Abramson Cancer Center; Nageatte Ibrahim, Elizabeth Cunningham, Merck; Peng Sun, Amy S. Kline, Heather Del Buono, Diane Opatt McDowell, GlaxoSmithKline, Philadelphia, PA; Donald Lawrence and Kiran Patel, Incyte Corporation, Wilmington, DE; and Keith T. Flaherty, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Adil Daud
- Georgina V. Long, Melanoma Institute Australia; The University of Sydney; Richard F. Kefford, Melanoma Institute Australia; The University of Sydney; Macquarie University, Sydney; Westmead Hospital, Westmead; Jonathan Cebon, Austin Health, Melbourne, Victoria, Australia; Jeffrey S. Weber and Ragini Kudchadkar, Moffitt Cancer Center, Tampa, FL; Jeffrey R. Infante and Howard A. Burris III, Sarah Cannon Research Institute/Tennessee Oncology; Kevin B. Kim, California Pacific Medical Center; Adil Daud, Alain Algazi, University of California, San Francisco, San Francisco; Omid Hamid, The Angeles Clinic and Research Institute, Los Angeles, CA; Rene Gonzalez, Karl Lewis, University of Colorado; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver, CO; Jeffrey A. Sosman, Igor Puzanov, Vanderbilt University Medical Center, Nashville, TN; Lynn Schuchter, University of Pennsylvania Abramson Cancer Center; Nageatte Ibrahim, Elizabeth Cunningham, Merck; Peng Sun, Amy S. Kline, Heather Del Buono, Diane Opatt McDowell, GlaxoSmithKline, Philadelphia, PA; Donald Lawrence and Kiran Patel, Incyte Corporation, Wilmington, DE; and Keith T. Flaherty, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Rene Gonzalez
- Georgina V. Long, Melanoma Institute Australia; The University of Sydney; Richard F. Kefford, Melanoma Institute Australia; The University of Sydney; Macquarie University, Sydney; Westmead Hospital, Westmead; Jonathan Cebon, Austin Health, Melbourne, Victoria, Australia; Jeffrey S. Weber and Ragini Kudchadkar, Moffitt Cancer Center, Tampa, FL; Jeffrey R. Infante and Howard A. Burris III, Sarah Cannon Research Institute/Tennessee Oncology; Kevin B. Kim, California Pacific Medical Center; Adil Daud, Alain Algazi, University of California, San Francisco, San Francisco; Omid Hamid, The Angeles Clinic and Research Institute, Los Angeles, CA; Rene Gonzalez, Karl Lewis, University of Colorado; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver, CO; Jeffrey A. Sosman, Igor Puzanov, Vanderbilt University Medical Center, Nashville, TN; Lynn Schuchter, University of Pennsylvania Abramson Cancer Center; Nageatte Ibrahim, Elizabeth Cunningham, Merck; Peng Sun, Amy S. Kline, Heather Del Buono, Diane Opatt McDowell, GlaxoSmithKline, Philadelphia, PA; Donald Lawrence and Kiran Patel, Incyte Corporation, Wilmington, DE; and Keith T. Flaherty, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Jeffrey A Sosman
- Georgina V. Long, Melanoma Institute Australia; The University of Sydney; Richard F. Kefford, Melanoma Institute Australia; The University of Sydney; Macquarie University, Sydney; Westmead Hospital, Westmead; Jonathan Cebon, Austin Health, Melbourne, Victoria, Australia; Jeffrey S. Weber and Ragini Kudchadkar, Moffitt Cancer Center, Tampa, FL; Jeffrey R. Infante and Howard A. Burris III, Sarah Cannon Research Institute/Tennessee Oncology; Kevin B. Kim, California Pacific Medical Center; Adil Daud, Alain Algazi, University of California, San Francisco, San Francisco; Omid Hamid, The Angeles Clinic and Research Institute, Los Angeles, CA; Rene Gonzalez, Karl Lewis, University of Colorado; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver, CO; Jeffrey A. Sosman, Igor Puzanov, Vanderbilt University Medical Center, Nashville, TN; Lynn Schuchter, University of Pennsylvania Abramson Cancer Center; Nageatte Ibrahim, Elizabeth Cunningham, Merck; Peng Sun, Amy S. Kline, Heather Del Buono, Diane Opatt McDowell, GlaxoSmithKline, Philadelphia, PA; Donald Lawrence and Kiran Patel, Incyte Corporation, Wilmington, DE; and Keith T. Flaherty, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Omid Hamid
- Georgina V. Long, Melanoma Institute Australia; The University of Sydney; Richard F. Kefford, Melanoma Institute Australia; The University of Sydney; Macquarie University, Sydney; Westmead Hospital, Westmead; Jonathan Cebon, Austin Health, Melbourne, Victoria, Australia; Jeffrey S. Weber and Ragini Kudchadkar, Moffitt Cancer Center, Tampa, FL; Jeffrey R. Infante and Howard A. Burris III, Sarah Cannon Research Institute/Tennessee Oncology; Kevin B. Kim, California Pacific Medical Center; Adil Daud, Alain Algazi, University of California, San Francisco, San Francisco; Omid Hamid, The Angeles Clinic and Research Institute, Los Angeles, CA; Rene Gonzalez, Karl Lewis, University of Colorado; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver, CO; Jeffrey A. Sosman, Igor Puzanov, Vanderbilt University Medical Center, Nashville, TN; Lynn Schuchter, University of Pennsylvania Abramson Cancer Center; Nageatte Ibrahim, Elizabeth Cunningham, Merck; Peng Sun, Amy S. Kline, Heather Del Buono, Diane Opatt McDowell, GlaxoSmithKline, Philadelphia, PA; Donald Lawrence and Kiran Patel, Incyte Corporation, Wilmington, DE; and Keith T. Flaherty, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Lynn Schuchter
- Georgina V. Long, Melanoma Institute Australia; The University of Sydney; Richard F. Kefford, Melanoma Institute Australia; The University of Sydney; Macquarie University, Sydney; Westmead Hospital, Westmead; Jonathan Cebon, Austin Health, Melbourne, Victoria, Australia; Jeffrey S. Weber and Ragini Kudchadkar, Moffitt Cancer Center, Tampa, FL; Jeffrey R. Infante and Howard A. Burris III, Sarah Cannon Research Institute/Tennessee Oncology; Kevin B. Kim, California Pacific Medical Center; Adil Daud, Alain Algazi, University of California, San Francisco, San Francisco; Omid Hamid, The Angeles Clinic and Research Institute, Los Angeles, CA; Rene Gonzalez, Karl Lewis, University of Colorado; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver, CO; Jeffrey A. Sosman, Igor Puzanov, Vanderbilt University Medical Center, Nashville, TN; Lynn Schuchter, University of Pennsylvania Abramson Cancer Center; Nageatte Ibrahim, Elizabeth Cunningham, Merck; Peng Sun, Amy S. Kline, Heather Del Buono, Diane Opatt McDowell, GlaxoSmithKline, Philadelphia, PA; Donald Lawrence and Kiran Patel, Incyte Corporation, Wilmington, DE; and Keith T. Flaherty, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Jonathan Cebon
- Georgina V. Long, Melanoma Institute Australia; The University of Sydney; Richard F. Kefford, Melanoma Institute Australia; The University of Sydney; Macquarie University, Sydney; Westmead Hospital, Westmead; Jonathan Cebon, Austin Health, Melbourne, Victoria, Australia; Jeffrey S. Weber and Ragini Kudchadkar, Moffitt Cancer Center, Tampa, FL; Jeffrey R. Infante and Howard A. Burris III, Sarah Cannon Research Institute/Tennessee Oncology; Kevin B. Kim, California Pacific Medical Center; Adil Daud, Alain Algazi, University of California, San Francisco, San Francisco; Omid Hamid, The Angeles Clinic and Research Institute, Los Angeles, CA; Rene Gonzalez, Karl Lewis, University of Colorado; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver, CO; Jeffrey A. Sosman, Igor Puzanov, Vanderbilt University Medical Center, Nashville, TN; Lynn Schuchter, University of Pennsylvania Abramson Cancer Center; Nageatte Ibrahim, Elizabeth Cunningham, Merck; Peng Sun, Amy S. Kline, Heather Del Buono, Diane Opatt McDowell, GlaxoSmithKline, Philadelphia, PA; Donald Lawrence and Kiran Patel, Incyte Corporation, Wilmington, DE; and Keith T. Flaherty, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Richard F Kefford
- Georgina V. Long, Melanoma Institute Australia; The University of Sydney; Richard F. Kefford, Melanoma Institute Australia; The University of Sydney; Macquarie University, Sydney; Westmead Hospital, Westmead; Jonathan Cebon, Austin Health, Melbourne, Victoria, Australia; Jeffrey S. Weber and Ragini Kudchadkar, Moffitt Cancer Center, Tampa, FL; Jeffrey R. Infante and Howard A. Burris III, Sarah Cannon Research Institute/Tennessee Oncology; Kevin B. Kim, California Pacific Medical Center; Adil Daud, Alain Algazi, University of California, San Francisco, San Francisco; Omid Hamid, The Angeles Clinic and Research Institute, Los Angeles, CA; Rene Gonzalez, Karl Lewis, University of Colorado; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver, CO; Jeffrey A. Sosman, Igor Puzanov, Vanderbilt University Medical Center, Nashville, TN; Lynn Schuchter, University of Pennsylvania Abramson Cancer Center; Nageatte Ibrahim, Elizabeth Cunningham, Merck; Peng Sun, Amy S. Kline, Heather Del Buono, Diane Opatt McDowell, GlaxoSmithKline, Philadelphia, PA; Donald Lawrence and Kiran Patel, Incyte Corporation, Wilmington, DE; and Keith T. Flaherty, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Donald Lawrence
- Georgina V. Long, Melanoma Institute Australia; The University of Sydney; Richard F. Kefford, Melanoma Institute Australia; The University of Sydney; Macquarie University, Sydney; Westmead Hospital, Westmead; Jonathan Cebon, Austin Health, Melbourne, Victoria, Australia; Jeffrey S. Weber and Ragini Kudchadkar, Moffitt Cancer Center, Tampa, FL; Jeffrey R. Infante and Howard A. Burris III, Sarah Cannon Research Institute/Tennessee Oncology; Kevin B. Kim, California Pacific Medical Center; Adil Daud, Alain Algazi, University of California, San Francisco, San Francisco; Omid Hamid, The Angeles Clinic and Research Institute, Los Angeles, CA; Rene Gonzalez, Karl Lewis, University of Colorado; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver, CO; Jeffrey A. Sosman, Igor Puzanov, Vanderbilt University Medical Center, Nashville, TN; Lynn Schuchter, University of Pennsylvania Abramson Cancer Center; Nageatte Ibrahim, Elizabeth Cunningham, Merck; Peng Sun, Amy S. Kline, Heather Del Buono, Diane Opatt McDowell, GlaxoSmithKline, Philadelphia, PA; Donald Lawrence and Kiran Patel, Incyte Corporation, Wilmington, DE; and Keith T. Flaherty, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Ragini Kudchadkar
- Georgina V. Long, Melanoma Institute Australia; The University of Sydney; Richard F. Kefford, Melanoma Institute Australia; The University of Sydney; Macquarie University, Sydney; Westmead Hospital, Westmead; Jonathan Cebon, Austin Health, Melbourne, Victoria, Australia; Jeffrey S. Weber and Ragini Kudchadkar, Moffitt Cancer Center, Tampa, FL; Jeffrey R. Infante and Howard A. Burris III, Sarah Cannon Research Institute/Tennessee Oncology; Kevin B. Kim, California Pacific Medical Center; Adil Daud, Alain Algazi, University of California, San Francisco, San Francisco; Omid Hamid, The Angeles Clinic and Research Institute, Los Angeles, CA; Rene Gonzalez, Karl Lewis, University of Colorado; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver, CO; Jeffrey A. Sosman, Igor Puzanov, Vanderbilt University Medical Center, Nashville, TN; Lynn Schuchter, University of Pennsylvania Abramson Cancer Center; Nageatte Ibrahim, Elizabeth Cunningham, Merck; Peng Sun, Amy S. Kline, Heather Del Buono, Diane Opatt McDowell, GlaxoSmithKline, Philadelphia, PA; Donald Lawrence and Kiran Patel, Incyte Corporation, Wilmington, DE; and Keith T. Flaherty, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Howard A Burris
- Georgina V. Long, Melanoma Institute Australia; The University of Sydney; Richard F. Kefford, Melanoma Institute Australia; The University of Sydney; Macquarie University, Sydney; Westmead Hospital, Westmead; Jonathan Cebon, Austin Health, Melbourne, Victoria, Australia; Jeffrey S. Weber and Ragini Kudchadkar, Moffitt Cancer Center, Tampa, FL; Jeffrey R. Infante and Howard A. Burris III, Sarah Cannon Research Institute/Tennessee Oncology; Kevin B. Kim, California Pacific Medical Center; Adil Daud, Alain Algazi, University of California, San Francisco, San Francisco; Omid Hamid, The Angeles Clinic and Research Institute, Los Angeles, CA; Rene Gonzalez, Karl Lewis, University of Colorado; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver, CO; Jeffrey A. Sosman, Igor Puzanov, Vanderbilt University Medical Center, Nashville, TN; Lynn Schuchter, University of Pennsylvania Abramson Cancer Center; Nageatte Ibrahim, Elizabeth Cunningham, Merck; Peng Sun, Amy S. Kline, Heather Del Buono, Diane Opatt McDowell, GlaxoSmithKline, Philadelphia, PA; Donald Lawrence and Kiran Patel, Incyte Corporation, Wilmington, DE; and Keith T. Flaherty, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Gerald S Falchook
- Georgina V. Long, Melanoma Institute Australia; The University of Sydney; Richard F. Kefford, Melanoma Institute Australia; The University of Sydney; Macquarie University, Sydney; Westmead Hospital, Westmead; Jonathan Cebon, Austin Health, Melbourne, Victoria, Australia; Jeffrey S. Weber and Ragini Kudchadkar, Moffitt Cancer Center, Tampa, FL; Jeffrey R. Infante and Howard A. Burris III, Sarah Cannon Research Institute/Tennessee Oncology; Kevin B. Kim, California Pacific Medical Center; Adil Daud, Alain Algazi, University of California, San Francisco, San Francisco; Omid Hamid, The Angeles Clinic and Research Institute, Los Angeles, CA; Rene Gonzalez, Karl Lewis, University of Colorado; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver, CO; Jeffrey A. Sosman, Igor Puzanov, Vanderbilt University Medical Center, Nashville, TN; Lynn Schuchter, University of Pennsylvania Abramson Cancer Center; Nageatte Ibrahim, Elizabeth Cunningham, Merck; Peng Sun, Amy S. Kline, Heather Del Buono, Diane Opatt McDowell, GlaxoSmithKline, Philadelphia, PA; Donald Lawrence and Kiran Patel, Incyte Corporation, Wilmington, DE; and Keith T. Flaherty, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Alain Algazi
- Georgina V. Long, Melanoma Institute Australia; The University of Sydney; Richard F. Kefford, Melanoma Institute Australia; The University of Sydney; Macquarie University, Sydney; Westmead Hospital, Westmead; Jonathan Cebon, Austin Health, Melbourne, Victoria, Australia; Jeffrey S. Weber and Ragini Kudchadkar, Moffitt Cancer Center, Tampa, FL; Jeffrey R. Infante and Howard A. Burris III, Sarah Cannon Research Institute/Tennessee Oncology; Kevin B. Kim, California Pacific Medical Center; Adil Daud, Alain Algazi, University of California, San Francisco, San Francisco; Omid Hamid, The Angeles Clinic and Research Institute, Los Angeles, CA; Rene Gonzalez, Karl Lewis, University of Colorado; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver, CO; Jeffrey A. Sosman, Igor Puzanov, Vanderbilt University Medical Center, Nashville, TN; Lynn Schuchter, University of Pennsylvania Abramson Cancer Center; Nageatte Ibrahim, Elizabeth Cunningham, Merck; Peng Sun, Amy S. Kline, Heather Del Buono, Diane Opatt McDowell, GlaxoSmithKline, Philadelphia, PA; Donald Lawrence and Kiran Patel, Incyte Corporation, Wilmington, DE; and Keith T. Flaherty, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Karl Lewis
- Georgina V. Long, Melanoma Institute Australia; The University of Sydney; Richard F. Kefford, Melanoma Institute Australia; The University of Sydney; Macquarie University, Sydney; Westmead Hospital, Westmead; Jonathan Cebon, Austin Health, Melbourne, Victoria, Australia; Jeffrey S. Weber and Ragini Kudchadkar, Moffitt Cancer Center, Tampa, FL; Jeffrey R. Infante and Howard A. Burris III, Sarah Cannon Research Institute/Tennessee Oncology; Kevin B. Kim, California Pacific Medical Center; Adil Daud, Alain Algazi, University of California, San Francisco, San Francisco; Omid Hamid, The Angeles Clinic and Research Institute, Los Angeles, CA; Rene Gonzalez, Karl Lewis, University of Colorado; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver, CO; Jeffrey A. Sosman, Igor Puzanov, Vanderbilt University Medical Center, Nashville, TN; Lynn Schuchter, University of Pennsylvania Abramson Cancer Center; Nageatte Ibrahim, Elizabeth Cunningham, Merck; Peng Sun, Amy S. Kline, Heather Del Buono, Diane Opatt McDowell, GlaxoSmithKline, Philadelphia, PA; Donald Lawrence and Kiran Patel, Incyte Corporation, Wilmington, DE; and Keith T. Flaherty, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Igor Puzanov
- Georgina V. Long, Melanoma Institute Australia; The University of Sydney; Richard F. Kefford, Melanoma Institute Australia; The University of Sydney; Macquarie University, Sydney; Westmead Hospital, Westmead; Jonathan Cebon, Austin Health, Melbourne, Victoria, Australia; Jeffrey S. Weber and Ragini Kudchadkar, Moffitt Cancer Center, Tampa, FL; Jeffrey R. Infante and Howard A. Burris III, Sarah Cannon Research Institute/Tennessee Oncology; Kevin B. Kim, California Pacific Medical Center; Adil Daud, Alain Algazi, University of California, San Francisco, San Francisco; Omid Hamid, The Angeles Clinic and Research Institute, Los Angeles, CA; Rene Gonzalez, Karl Lewis, University of Colorado; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver, CO; Jeffrey A. Sosman, Igor Puzanov, Vanderbilt University Medical Center, Nashville, TN; Lynn Schuchter, University of Pennsylvania Abramson Cancer Center; Nageatte Ibrahim, Elizabeth Cunningham, Merck; Peng Sun, Amy S. Kline, Heather Del Buono, Diane Opatt McDowell, GlaxoSmithKline, Philadelphia, PA; Donald Lawrence and Kiran Patel, Incyte Corporation, Wilmington, DE; and Keith T. Flaherty, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Nageatte Ibrahim
- Georgina V. Long, Melanoma Institute Australia; The University of Sydney; Richard F. Kefford, Melanoma Institute Australia; The University of Sydney; Macquarie University, Sydney; Westmead Hospital, Westmead; Jonathan Cebon, Austin Health, Melbourne, Victoria, Australia; Jeffrey S. Weber and Ragini Kudchadkar, Moffitt Cancer Center, Tampa, FL; Jeffrey R. Infante and Howard A. Burris III, Sarah Cannon Research Institute/Tennessee Oncology; Kevin B. Kim, California Pacific Medical Center; Adil Daud, Alain Algazi, University of California, San Francisco, San Francisco; Omid Hamid, The Angeles Clinic and Research Institute, Los Angeles, CA; Rene Gonzalez, Karl Lewis, University of Colorado; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver, CO; Jeffrey A. Sosman, Igor Puzanov, Vanderbilt University Medical Center, Nashville, TN; Lynn Schuchter, University of Pennsylvania Abramson Cancer Center; Nageatte Ibrahim, Elizabeth Cunningham, Merck; Peng Sun, Amy S. Kline, Heather Del Buono, Diane Opatt McDowell, GlaxoSmithKline, Philadelphia, PA; Donald Lawrence and Kiran Patel, Incyte Corporation, Wilmington, DE; and Keith T. Flaherty, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Peng Sun
- Georgina V. Long, Melanoma Institute Australia; The University of Sydney; Richard F. Kefford, Melanoma Institute Australia; The University of Sydney; Macquarie University, Sydney; Westmead Hospital, Westmead; Jonathan Cebon, Austin Health, Melbourne, Victoria, Australia; Jeffrey S. Weber and Ragini Kudchadkar, Moffitt Cancer Center, Tampa, FL; Jeffrey R. Infante and Howard A. Burris III, Sarah Cannon Research Institute/Tennessee Oncology; Kevin B. Kim, California Pacific Medical Center; Adil Daud, Alain Algazi, University of California, San Francisco, San Francisco; Omid Hamid, The Angeles Clinic and Research Institute, Los Angeles, CA; Rene Gonzalez, Karl Lewis, University of Colorado; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver, CO; Jeffrey A. Sosman, Igor Puzanov, Vanderbilt University Medical Center, Nashville, TN; Lynn Schuchter, University of Pennsylvania Abramson Cancer Center; Nageatte Ibrahim, Elizabeth Cunningham, Merck; Peng Sun, Amy S. Kline, Heather Del Buono, Diane Opatt McDowell, GlaxoSmithKline, Philadelphia, PA; Donald Lawrence and Kiran Patel, Incyte Corporation, Wilmington, DE; and Keith T. Flaherty, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Elizabeth Cunningham
- Georgina V. Long, Melanoma Institute Australia; The University of Sydney; Richard F. Kefford, Melanoma Institute Australia; The University of Sydney; Macquarie University, Sydney; Westmead Hospital, Westmead; Jonathan Cebon, Austin Health, Melbourne, Victoria, Australia; Jeffrey S. Weber and Ragini Kudchadkar, Moffitt Cancer Center, Tampa, FL; Jeffrey R. Infante and Howard A. Burris III, Sarah Cannon Research Institute/Tennessee Oncology; Kevin B. Kim, California Pacific Medical Center; Adil Daud, Alain Algazi, University of California, San Francisco, San Francisco; Omid Hamid, The Angeles Clinic and Research Institute, Los Angeles, CA; Rene Gonzalez, Karl Lewis, University of Colorado; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver, CO; Jeffrey A. Sosman, Igor Puzanov, Vanderbilt University Medical Center, Nashville, TN; Lynn Schuchter, University of Pennsylvania Abramson Cancer Center; Nageatte Ibrahim, Elizabeth Cunningham, Merck; Peng Sun, Amy S. Kline, Heather Del Buono, Diane Opatt McDowell, GlaxoSmithKline, Philadelphia, PA; Donald Lawrence and Kiran Patel, Incyte Corporation, Wilmington, DE; and Keith T. Flaherty, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Amy S Kline
- Georgina V. Long, Melanoma Institute Australia; The University of Sydney; Richard F. Kefford, Melanoma Institute Australia; The University of Sydney; Macquarie University, Sydney; Westmead Hospital, Westmead; Jonathan Cebon, Austin Health, Melbourne, Victoria, Australia; Jeffrey S. Weber and Ragini Kudchadkar, Moffitt Cancer Center, Tampa, FL; Jeffrey R. Infante and Howard A. Burris III, Sarah Cannon Research Institute/Tennessee Oncology; Kevin B. Kim, California Pacific Medical Center; Adil Daud, Alain Algazi, University of California, San Francisco, San Francisco; Omid Hamid, The Angeles Clinic and Research Institute, Los Angeles, CA; Rene Gonzalez, Karl Lewis, University of Colorado; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver, CO; Jeffrey A. Sosman, Igor Puzanov, Vanderbilt University Medical Center, Nashville, TN; Lynn Schuchter, University of Pennsylvania Abramson Cancer Center; Nageatte Ibrahim, Elizabeth Cunningham, Merck; Peng Sun, Amy S. Kline, Heather Del Buono, Diane Opatt McDowell, GlaxoSmithKline, Philadelphia, PA; Donald Lawrence and Kiran Patel, Incyte Corporation, Wilmington, DE; and Keith T. Flaherty, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Heather Del Buono
- Georgina V. Long, Melanoma Institute Australia; The University of Sydney; Richard F. Kefford, Melanoma Institute Australia; The University of Sydney; Macquarie University, Sydney; Westmead Hospital, Westmead; Jonathan Cebon, Austin Health, Melbourne, Victoria, Australia; Jeffrey S. Weber and Ragini Kudchadkar, Moffitt Cancer Center, Tampa, FL; Jeffrey R. Infante and Howard A. Burris III, Sarah Cannon Research Institute/Tennessee Oncology; Kevin B. Kim, California Pacific Medical Center; Adil Daud, Alain Algazi, University of California, San Francisco, San Francisco; Omid Hamid, The Angeles Clinic and Research Institute, Los Angeles, CA; Rene Gonzalez, Karl Lewis, University of Colorado; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver, CO; Jeffrey A. Sosman, Igor Puzanov, Vanderbilt University Medical Center, Nashville, TN; Lynn Schuchter, University of Pennsylvania Abramson Cancer Center; Nageatte Ibrahim, Elizabeth Cunningham, Merck; Peng Sun, Amy S. Kline, Heather Del Buono, Diane Opatt McDowell, GlaxoSmithKline, Philadelphia, PA; Donald Lawrence and Kiran Patel, Incyte Corporation, Wilmington, DE; and Keith T. Flaherty, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Diane Opatt McDowell
- Georgina V. Long, Melanoma Institute Australia; The University of Sydney; Richard F. Kefford, Melanoma Institute Australia; The University of Sydney; Macquarie University, Sydney; Westmead Hospital, Westmead; Jonathan Cebon, Austin Health, Melbourne, Victoria, Australia; Jeffrey S. Weber and Ragini Kudchadkar, Moffitt Cancer Center, Tampa, FL; Jeffrey R. Infante and Howard A. Burris III, Sarah Cannon Research Institute/Tennessee Oncology; Kevin B. Kim, California Pacific Medical Center; Adil Daud, Alain Algazi, University of California, San Francisco, San Francisco; Omid Hamid, The Angeles Clinic and Research Institute, Los Angeles, CA; Rene Gonzalez, Karl Lewis, University of Colorado; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver, CO; Jeffrey A. Sosman, Igor Puzanov, Vanderbilt University Medical Center, Nashville, TN; Lynn Schuchter, University of Pennsylvania Abramson Cancer Center; Nageatte Ibrahim, Elizabeth Cunningham, Merck; Peng Sun, Amy S. Kline, Heather Del Buono, Diane Opatt McDowell, GlaxoSmithKline, Philadelphia, PA; Donald Lawrence and Kiran Patel, Incyte Corporation, Wilmington, DE; and Keith T. Flaherty, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Kiran Patel
- Georgina V. Long, Melanoma Institute Australia; The University of Sydney; Richard F. Kefford, Melanoma Institute Australia; The University of Sydney; Macquarie University, Sydney; Westmead Hospital, Westmead; Jonathan Cebon, Austin Health, Melbourne, Victoria, Australia; Jeffrey S. Weber and Ragini Kudchadkar, Moffitt Cancer Center, Tampa, FL; Jeffrey R. Infante and Howard A. Burris III, Sarah Cannon Research Institute/Tennessee Oncology; Kevin B. Kim, California Pacific Medical Center; Adil Daud, Alain Algazi, University of California, San Francisco, San Francisco; Omid Hamid, The Angeles Clinic and Research Institute, Los Angeles, CA; Rene Gonzalez, Karl Lewis, University of Colorado; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver, CO; Jeffrey A. Sosman, Igor Puzanov, Vanderbilt University Medical Center, Nashville, TN; Lynn Schuchter, University of Pennsylvania Abramson Cancer Center; Nageatte Ibrahim, Elizabeth Cunningham, Merck; Peng Sun, Amy S. Kline, Heather Del Buono, Diane Opatt McDowell, GlaxoSmithKline, Philadelphia, PA; Donald Lawrence and Kiran Patel, Incyte Corporation, Wilmington, DE; and Keith T. Flaherty, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Keith T Flaherty
- Georgina V. Long, Melanoma Institute Australia; The University of Sydney; Richard F. Kefford, Melanoma Institute Australia; The University of Sydney; Macquarie University, Sydney; Westmead Hospital, Westmead; Jonathan Cebon, Austin Health, Melbourne, Victoria, Australia; Jeffrey S. Weber and Ragini Kudchadkar, Moffitt Cancer Center, Tampa, FL; Jeffrey R. Infante and Howard A. Burris III, Sarah Cannon Research Institute/Tennessee Oncology; Kevin B. Kim, California Pacific Medical Center; Adil Daud, Alain Algazi, University of California, San Francisco, San Francisco; Omid Hamid, The Angeles Clinic and Research Institute, Los Angeles, CA; Rene Gonzalez, Karl Lewis, University of Colorado; Gerald S. Falchook, Sarah Cannon Research Institute at HealthONE, Denver, CO; Jeffrey A. Sosman, Igor Puzanov, Vanderbilt University Medical Center, Nashville, TN; Lynn Schuchter, University of Pennsylvania Abramson Cancer Center; Nageatte Ibrahim, Elizabeth Cunningham, Merck; Peng Sun, Amy S. Kline, Heather Del Buono, Diane Opatt McDowell, GlaxoSmithKline, Philadelphia, PA; Donald Lawrence and Kiran Patel, Incyte Corporation, Wilmington, DE; and Keith T. Flaherty, Massachusetts General Hospital Cancer Center, Boston, MA
| |
Collapse
|
1777
|
Meyer N. [What's new in oncodermatology in 2015?]. Ann Dermatol Venereol 2016; 142 Suppl 3:S36-48. [PMID: 26792413 DOI: 10.1016/s0151-9638(16)30005-9] [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: 11/28/2022]
Abstract
Oncodermatology is the topic of constant innovation since 5 years. These innovations have drammatically modified the prognostic of skin cancers and induced a change in the paradigm that drives the whole oncology. Once again in 2015, melanoma is the key topic of scientific communication, with promising results. However, unmet clinical needs must be kept in light. Some diseases, such as squamous cell carcinoma or cutaneous lymphoma remain poorly evaluated. This article was designed as a review of litterature. All keywords of oncodermatology were searched in the main journals of dermatology, oncology and internal medicine. Despite methodologic restrictions, the number of articles imposed a choice based on opinion. The final goal was to transmit author's enthousiasm and the most important results.
Collapse
Affiliation(s)
- N Meyer
- Dermatologie, université Paul-Sabatier - Toulouse III et Institut Universitaire du Cancer de Toulouse, 1, rue Irène-Joliot- Curie, 31059 Toulouse Cedex 9 ; INSERM UMR 1037, CRCT, Toulouse, France.
| |
Collapse
|
1778
|
Diem S, Kasenda B, Spain L, Martin-Liberal J, Marconcini R, Gore M, Larkin J. Serum lactate dehydrogenase as an early marker for outcome in patients treated with anti-PD-1 therapy in metastatic melanoma. Br J Cancer 2016; 114:256-61. [PMID: 26794281 PMCID: PMC4742588 DOI: 10.1038/bjc.2015.467] [Citation(s) in RCA: 244] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Revised: 11/12/2015] [Accepted: 12/07/2015] [Indexed: 01/20/2023] Open
Abstract
Background: Treatment with programmed death receptor-1 (PD-1) antibodies is associated with high response rates in patients with advanced melanoma. Reliable markers for early response and outcome are still sparse. Methods: We evaluated 66 consecutive patients with advanced/metastatic melanoma treated with nivolumab or pembrolizumab between 2013 and 2014. The main objectives of this study were to investigate whether, first, serum lactate dehydrogenase (LDH) at baseline (normal vs above the upper limit of normal) correlates with overall survival (OS), and, second, whether the change of LDH during treatment predicts response before the first scan and OS in patients with an elevated baseline LDH. Results: After a median follow-up of 9 months, patients with an elevated baseline LDH (N=34) had a significantly shorter OS compared with patients with normal LDH (N=32; 6-month OS: 60.8% vs 81.6% and 12-month OS: 44.2% vs 71.5% (log-rank P=0.0292). In those 34 patients with elevated baseline LDH, the relative change during treatment was significantly associated with an objective response on the first scan: the 11 (32%) patients with partial remission had a mean reduction of −27.3% from elevated baseline LDH. In contrast, patients with progressive disease (N=15) had a mean increase of +39%. Patients with a relative increase over 10% from elevated baseline LDH had a significantly shorter OS compared with patients with ⩽10% change (4.3 vs 15.7 months, log-rank P<0.00623). Conclusions: LDH could be a useful marker at baseline and during treatment to predict early response or progression in patients with advanced melanoma who receive anti-PD-1 therapy.
Collapse
Affiliation(s)
- S Diem
- Department of Medical Oncology, Royal Marsden Hospital NHS Foundation Trust, Fulham Road, London SW36JJ, UK
| | - B Kasenda
- Department of Medical Oncology, Royal Marsden Hospital NHS Foundation Trust, Fulham Road, London SW36JJ, UK
| | - L Spain
- Department of Medical Oncology, Royal Marsden Hospital NHS Foundation Trust, Fulham Road, London SW36JJ, UK
| | - J Martin-Liberal
- Department of Medical Oncology, Royal Marsden Hospital NHS Foundation Trust, Fulham Road, London SW36JJ, UK.,Department of Medical Oncology, Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology (VHIO), Pg Vall d'Hebron, 119-129, 08035 Barcelona, Spain
| | - R Marconcini
- Department of Medical Oncology, Santa Chiara Hospital, via Roma 67, 56100 Pisa, Italy
| | - M Gore
- Department of Medical Oncology, Royal Marsden Hospital NHS Foundation Trust, Fulham Road, London SW36JJ, UK
| | - J Larkin
- Department of Medical Oncology, Royal Marsden Hospital NHS Foundation Trust, Fulham Road, London SW36JJ, UK
| |
Collapse
|
1779
|
Hovelson DH, McDaniel AS, Cani AK, Johnson B, Rhodes K, Williams PD, Bandla S, Bien G, Choppa P, Hyland F, Gottimukkala R, Liu G, Manivannan M, Schageman J, Ballesteros-Villagrana E, Grasso CS, Quist MJ, Yadati V, Amin A, Siddiqui J, Betz BL, Knudsen KE, Cooney KA, Feng FY, Roh MH, Nelson PS, Liu CJ, Beer DG, Wyngaard P, Chinnaiyan AM, Sadis S, Rhodes DR, Tomlins SA. Development and validation of a scalable next-generation sequencing system for assessing relevant somatic variants in solid tumors. Neoplasia 2016; 17:385-99. [PMID: 25925381 PMCID: PMC4415141 DOI: 10.1016/j.neo.2015.03.004] [Citation(s) in RCA: 176] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 03/27/2015] [Indexed: 12/21/2022] Open
Abstract
Next-generation sequencing (NGS) has enabled genome-wide personalized oncology efforts at centers and companies with the specialty expertise and infrastructure required to identify and prioritize actionable variants. Such approaches are not scalable, preventing widespread adoption. Likewise, most targeted NGS approaches fail to assess key relevant genomic alteration classes. To address these challenges, we predefined the catalog of relevant solid tumor somatic genome variants (gain-of-function or loss-of-function mutations, high-level copy number alterations, and gene fusions) through comprehensive bioinformatics analysis of >700,000 samples. To detect these variants, we developed the Oncomine Comprehensive Panel (OCP), an integrative NGS-based assay [compatible with < 20 ng of DNA/RNA from formalin-fixed paraffin-embedded (FFPE) tissues], coupled with an informatics pipeline to specifically identify relevant predefined variants and created a knowledge base of related potential treatments, current practice guidelines, and open clinical trials. We validated OCP using molecular standards and more than 300 FFPE tumor samples, achieving >95% accuracy for KRAS, epidermal growth factor receptor, and BRAF mutation detection as well as for ALK and TMPRSS2:ERG gene fusions. Associating positive variants with potential targeted treatments demonstrated that 6% to 42% of profiled samples (depending on cancer type) harbored alterations beyond routine molecular testing that were associated with approved or guideline-referenced therapies. As a translational research tool, OCP identified adaptive CTNNB1 amplifications/mutations in treated prostate cancers. Through predefining somatic variants in solid tumors and compiling associated potential treatment strategies, OCP represents a simplified, broadly applicable targeted NGS system with the potential to advance precision oncology efforts.
Collapse
Affiliation(s)
- Daniel H Hovelson
- Michigan Center for Translational Pathology, Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Andrew S McDaniel
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Andi K Cani
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | | | - Kate Rhodes
- Thermo Fisher Scientific, Ann Arbor, MI, USA
| | | | | | | | - Paul Choppa
- Thermo Fisher Scientific, Ann Arbor, MI, USA
| | | | | | - Guoying Liu
- Thermo Fisher Scientific, Ann Arbor, MI, USA
| | | | | | | | - Catherine S Grasso
- Department of Pathology, Oregon Health and Sciences University, Portland, OR, USA
| | - Michael J Quist
- Department of Pathology, Oregon Health and Sciences University, Portland, OR, USA
| | - Venkata Yadati
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Anmol Amin
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Javed Siddiqui
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Bryan L Betz
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Karen E Knudsen
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA; Department of Urology, Thomas Jefferson University, Philadelphia, PA, USA; Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Kathleen A Cooney
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA; Department of Urology, University of Michigan Medical School, Ann Arbor, MI, USA; Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Felix Y Feng
- Department of Radiation Oncology, University of Michigan Medical School, Ann Arbor, MI, USA; Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Michael H Roh
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Peter S Nelson
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, USA; Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Chia-Jen Liu
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - David G Beer
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA; Michigan Center for Translational Pathology, Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI, USA
| | | | - Arul M Chinnaiyan
- Michigan Center for Translational Pathology, Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI, USA; Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA; Department of Urology, University of Michigan Medical School, Ann Arbor, MI, USA; Michigan Center for Translational Pathology, Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI, USA; Howard Hughes Medical Institute, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Seth Sadis
- Thermo Fisher Scientific, Ann Arbor, MI, USA
| | - Daniel R Rhodes
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA; Thermo Fisher Scientific, Ann Arbor, MI, USA
| | - Scott A Tomlins
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA; Department of Urology, University of Michigan Medical School, Ann Arbor, MI, USA; Michigan Center for Translational Pathology, Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
1780
|
Martens A, Wistuba-Hamprecht K, Geukes Foppen M, Yuan J, Postow MA, Wong P, Romano E, Khammari A, Dreno B, Capone M, Ascierto PA, Di Giacomo AM, Maio M, Schilling B, Sucker A, Schadendorf D, Hassel JC, Eigentler TK, Martus P, Wolchok JD, Blank C, Pawelec G, Garbe C, Weide B. Baseline Peripheral Blood Biomarkers Associated with Clinical Outcome of Advanced Melanoma Patients Treated with Ipilimumab. Clin Cancer Res 2016; 22:2908-18. [PMID: 26787752 DOI: 10.1158/1078-0432.ccr-15-2412] [Citation(s) in RCA: 421] [Impact Index Per Article: 46.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 01/04/2016] [Indexed: 12/21/2022]
Abstract
PURPOSE To identify baseline peripheral blood biomarkers associated with clinical outcome following ipilimumab treatment in advanced melanoma patients. EXPERIMENTAL DESIGN Frequencies of myeloid-derived suppressor cells (MDSC) and regulatory T cells (Treg), serum lactate dehydrogenase (LDH), routine blood counts, and clinical characteristics were assessed in 209 patients. Endpoints were overall survival (OS) and best overall response. Statistical calculations were done by Kaplan-Meier and Cox regression analysis, including calibration and discrimination by C-statistics. RESULTS Low baseline LDH, absolute monocyte counts (AMC), Lin(-)CD14(+)HLA-DR(-/low)-MDSC frequencies, and high absolute eosinophil counts (AEC), relative lymphocyte counts (RLC), and CD4(+)CD25(+)FoxP3(+)-Treg frequencies were significantly associated with better survival, and were considered in a combination model. Patients (43.5%) presenting with the best biomarker signature had a 30% response rate and median survival of 16 months. In contrast, patients with the worst biomarkers (27.5%) had only a 3% response rate and median survival of 4 months. The occurrence of adverse events correlated with neither baseline biomarker signatures nor the clinical benefit of ipilimumab. In another model, limited to the routine parameters LDH, AMC, AEC, and RLC, the number of favorable factors (4 vs. 3 vs. 2-0) was also associated with OS (P < 0.001 for all pairwise comparisons) in the main study and additionally in an independent validation cohort. CONCLUSIONS A baseline signature of low LDH, AMC, and MDSCs as well as high AEC, Tregs, and RLC is associated with favorable outcome following ipilimumab. Prospective investigation of the predictive impact of these markers following ipilimumab and other treatments, e.g., PD-1 antibodies, is warranted. Clin Cancer Res; 22(12); 2908-18. ©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
| | - Emanuela Romano
- Department of Oncology, Service of Medical Oncology, Research Unit 932, Institut Curie, Paris, France
| | - Amir Khammari
- Department of Oncodermatology, INSERM Research Unit 892, University Hospital, Nantes, France
| | - Brigitte Dreno
- Department of Oncodermatology, INSERM Research Unit 892, University Hospital, Nantes, France
| | | | | | | | - Michele Maio
- Division of Medical Oncology and Immunotherapy, University Hospital of Siena, Italy
| | - 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
| | - Jessica C Hassel
- German Cancer Consortium (DKTK), Heidelberg, Germany. Department of Dermatology and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
| | | | - Peter Martus
- Departments of Clinical Epidemiology and Applied Biostatistics, University of Tübingen, Tübingen, Germany
| | - Jedd D Wolchok
- Memorial Sloan Kettering Cancer Center, New York, New York. Weill Cornell Medical College, New York, New York
| | - Christian Blank
- The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Graham Pawelec
- Department of Internal Medicine II, University Medical Center, Tübingen, Germany
| | - Claus Garbe
- Department of Dermatology, University Medical Center, Tübingen, Germany
| | - Benjamin Weide
- Department of Dermatology, University Medical Center, Tübingen, Germany. Department of Immunology, University of Tübingen, Tübingen, Germany.
| |
Collapse
|
1781
|
Wanchoo R, Jhaveri KD, Deray G, Launay-Vacher V. Renal effects of BRAF inhibitors: a systematic review by the Cancer and the Kidney International Network. Clin Kidney J 2016; 9:245-51. [PMID: 26985376 PMCID: PMC4792624 DOI: 10.1093/ckj/sfv149] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 12/03/2015] [Indexed: 12/11/2022] Open
Abstract
Advanced melanoma has been traditionally unresponsive to standard chemotherapy agents and used to have a dismal prognosis. Genetically targeted small-molecule inhibitors of the oncogenic BRAF V600 mutation or a downstream signaling partner (MEK mitogen-activated protein kinase) are effective treatment options for the 40–50% of melanomas that harbor mutations in BRAF. Selective BRAF and MEK inhibitors induce frequent and dramatic objective responses and markedly improve survival compared with cytotoxic chemotherapy. In the past decade after discovery of this mutation, drugs such as vemurafenib and dabrafenib have been approved by the US Food and Drug Administration (FDA) and the European Medicines Agency for the treatment of V600-mutated melanomas. While the initial trials did not signal any renal toxicities with the BRAF inhibitors, recent case reports, case series and FDA adverse reporting systems have uncovered significant nephrotoxicities with these agents. In this article, we systematically review the nephrotoxicities of these agents. Based on recently published data, it appears that there are lower rates of kidney disease and cutaneous lesions seen with dabrafenib compared with vemurafenib. The pathology reported in the few kidney biopsies done so far are suggestive of tubulo interstitial damage with an acute and chronic component. Electrolyte disorders such as hypokalemia, hyponatremia and hypophosphatemia have been reported as well. Routine monitoring of serum creatinine and electrolytes and calculation of glomerular filtration rate prior to the first administration when treating with dabrafenib and vemurafenib are essential.
Collapse
Affiliation(s)
- Rimda Wanchoo
- Division of Kidney Diseases and Hypertension , North Shore University Hospital and Long Island Jewish Medical Center, Hofstra NSLIJ School of Medicine , Great Neck, NY , USA
| | - Kenar D Jhaveri
- Division of Kidney Diseases and Hypertension , North Shore University Hospital and Long Island Jewish Medical Center, Hofstra NSLIJ School of Medicine , Great Neck, NY , USA
| | - Gilbert Deray
- Nephrology Department , Pitié-Salpêtrière University Hospital , Paris , France
| | - Vincent Launay-Vacher
- Nephrology Department, Pitié-Salpêtrière University Hospital, Paris, France; Service ICAR, Pitié-Salpêtrière University Hospital, Paris, France
| |
Collapse
|
1782
|
The molecular profile of metastatic melanoma in Australia. Pathology 2016; 48:188-93. [PMID: 27020391 DOI: 10.1016/j.pathol.2015.12.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Accepted: 11/26/2015] [Indexed: 11/24/2022]
Abstract
Targeted therapy directed at driver oncogenic mutations offers an effective treatment option for select patients with metastatic melanoma. The aim of this study was to assess the prevalence of clinically significant somatic mutations, specifically BRAF, NRAS and KIT, in a large cohort of Australian patients with metastatic melanoma. We performed a cross-sectional cohort study of consecutive patients with American Joint Committee on Cancer (AJCC) stage IIIc unresectable or stage IV melanoma managed at Melanoma Institute Australia, and affiliated sites, that underwent molecular testing between 22 June 2009 and 19 July 2013. Additionally, we examined the change in BRAF testing methodology and patient population over time, and how this influenced the prevalence of mutations. A total of 767 molecular tests were conducted for 733 patients. BRAF V600 mutation testing was performed for 713 patients (97.2%), with an overall mutation prevalence of 37.7% (269/713); 74.3% (200/269) were the V600E genotype and 22.3% (60/269) V600K. The BRAF mutation prevalence and proportion of BRAF V600E and V600K genotypes varied across the study period, as did testing methodology and the median age of the cohorts. Of 222 patients who underwent NRAS testing, 58 (26.1%) had a mutation identified. The overall prevalence of KIT mutations was 3.7% (11/296). In Australia the prevalence of BRAF mutations is lower than initially reported, although this remains the most common mutation identified in metastatic melanoma and an important therapeutic target. NRAS mutations are more prevalent than initially described; however, other mutations reported in melanoma, including KIT, are rare in an unselected population of patients.
Collapse
|
1783
|
Singh BP, Salama AKS. Updates in Therapy for Advanced Melanoma. Cancers (Basel) 2016; 8:E17. [PMID: 26784231 PMCID: PMC4728464 DOI: 10.3390/cancers8010017] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Revised: 01/04/2016] [Accepted: 01/07/2016] [Indexed: 01/15/2023] Open
Abstract
Cutaneous melanoma is one of the most aggressive forms of skin cancer, and is correlated with a large proportion of skin cancer-related deaths. Therapy for cutaneous melanoma has advanced greatly through careful identification of therapeutic targets and the development of novel immunotherapeutic approaches. The identification of BRAF as well as other driver mutations, have allowed for a specialized approach to treatment. In addition, immune checkpoint inhibition has dramatically changed the treatment landscape over the past 5-10 years. The successful targeting of CTLA-4, as well as PD-1/PD-L1, has been translated into meaningful clinical benefit for patients, with multiple other potential agents in development. Systemic therapy for cutaneous melanoma is becoming more nuanced and often takes a multifaceted strategy. This review aims to discuss the benefits and limitations of current therapies in systemic melanoma treatment as well as areas of future development.
Collapse
Affiliation(s)
- Bhavana P Singh
- Department of Internal Medicine, Duke University Medical Center, Durham, NC 27710, USA.
| | - April K S Salama
- Division of Medical Oncology, Department of Internal Medicine, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
1784
|
Harmon CM, Brown N. Langerhans Cell Histiocytosis: A Clinicopathologic Review and Molecular Pathogenetic Update. Arch Pathol Lab Med 2016; 139:1211-4. [PMID: 26414464 DOI: 10.5858/arpa.2015-0199-ra] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Langerhans cell histiocytosis (LCH) comprises a wide spectrum of clinical disorders that have in common a proliferation of Langerhans-type cells with characteristic morphologic, immunophenotypic, and ultrastructural features. In part because of the diverse clinical manifestations of LCH, there has long been controversy over whether LCH is best considered a reactive process or a neoplasm. Herein, we discuss the clinical and pathologic features of LCH, including recent advances in the understanding of the molecular pathogenesis of this disease that support its categorization as a neoplasm. We also review the implications that these recently described molecular characteristics may have on risk stratification and treatment of LCH.
Collapse
Affiliation(s)
| | - Noah Brown
- From the Department of Pathology, University of Michigan Hospital and Health Systems, Ann Arbor
| |
Collapse
|
1785
|
Meador CB, Pao W. Old Habits Die Hard: Addiction of BRAF-Mutant Cancer Cells to MAP Kinase Signaling. Cancer Discov 2016; 5:348-50. [PMID: 25847954 DOI: 10.1158/2159-8290.cd-15-0221] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Dual and triple combination therapies with RAF inhibitors plus other targeted agents have demonstrated promising clinical utility in BRAFV600-mutant solid tumors. However, despite vertical inhibition at multiple nodes on the MAPK signaling pathway, resistant tumors emerge. Ahronian and colleagues show that in BRAF-mutant colorectal cancer, resistance involves reactivation of RAS/RAF/MEK/ERK signaling and may be overcome by newly emerging ERK inhibitors.
Collapse
Affiliation(s)
- Catherine B Meador
- Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - William Pao
- Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, Tennessee. Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee.
| |
Collapse
|
1786
|
Comparison of dabrafenib and trametinib combination therapy with vemurafenib monotherapy on health-related quality of life in patients with unresectable or metastatic cutaneous BRAF Val600-mutation-positive melanoma (COMBI-v): results of a phase 3, open-label, randomised trial. Lancet Oncol 2016; 16:1389-98. [PMID: 26433819 DOI: 10.1016/s1470-2045(15)00087-x] [Citation(s) in RCA: 196] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Revised: 06/15/2015] [Accepted: 06/15/2015] [Indexed: 12/22/2022]
Abstract
BACKGROUND In the COMBI-v trial, patients with previously untreated BRAF Val600Glu or Val600Lys mutant unresectable or metastatic melanoma who were treated with the combination of dabrafenib and trametinib had significantly longer overall and progression-free survival than those treated with vemurafenib alone. Here, we present the effects of treatments on health-related quality of life (HRQoL), an exploratory endpoint in the COMBI-v study. METHODS COMBI-v was an open-label, randomised phase 3 study in which 704 patients with metastatic melanoma with a BRAF Val600 mutation were randomly assigned (1:1) by an interactive voice response system to receive either a combination of dabrafenib (150 mg twice-daily) and trametinib (2 mg once-daily) or vemurafenib monotherapy (960 mg twice-daily) orally as first-line therapy. The primary endpoint was overall survival. In this pre-specified exploratory analysis, we prospectively assessed HRQoL in the intention-to-treat population with the European Organisation for Research and Treatment of Cancer quality of life (EORTC QLQ-C30), EuroQoL-5D (EQ-5D), and Melanoma Subscale of the Functional Assessment of Cancer Therapy-Melanoma (FACT-M), completed at baseline, during study treatment, at disease progression, and after progression. We used a mixed-model, repeated measures ANCOVA to assess differences in mean scores between groups with baseline score as covariate; all p-values are descriptive. The COMBI-v trial is registered with ClinicalTrials.gov, number NCT01597908, and is ongoing for the primary endpoint, but is not recruiting patients. FINDINGS From June 4, 2012, to Oct 7, 2013, 1645 patients at 193 centres worldwide were screened for eligibility, and 704 patients were randomly assigned to dabrafenib plus trametinib (n=352) or vemurafenib (n=352). Questionnaire completion rates for both groups were high (>95% at baseline, >80% at follow-up assessments, and >70% at disease progression) with similar HRQoL and symptom scores reported at baseline in both treatment groups for all questionnaires. Differences in mean scores between treatment groups were significant and clinically meaningful in favour of the combination compared with vemurafenib monotherapy for most domains across all three questionnaires during study treatment and at disease progression, including EORTC QLQ-C30 global health (7·92, 7·62, 6·86, 7·47, 5·16, 7·56, and 7·57 at weeks 8, 16, 24, 32, 40, 48, and disease progression, respectively; p<0·001 for all assessments except p=0·005 at week 40), EORTC QLQ-C30 pain (-13·20, -8·05, -8·82, -12·69, -12·46, -11·41, and -10·57 at weeks 8, 16, 24, 32, 40, 48, and disease progression, respectively; all p<0·001), EQ-5D thermometer scores (7·96, 8·05, 6·83, 11·53, 7·41, 9·08, and 10·51 at weeks 8, 16, 24, 32, 40, 48, and disease progression, respectively; p<0·001 for all assessments except p=0·006 at week 32), and FACT-M Melanoma Subscale score (3·62, 2·93, 2·45, 3·39, 2·85, 3·00, and 3·68 at weeks 8, 16, 24, 32, 40, 48, and disease progression, respectively; all p<0·001). INTERPRETATION From the patient's perspective, which integrates not only survival advantage but also disease-associated and adverse-event-associated symptoms, treatment with the combination of a BRAF inhibitor plus a MEK inhibitor (dabrafenib plus trametinib) adds a clear benefit over monotherapy with the BRAF inhibitor vemurafenib and supports the combination therapy as standard of care in this population.
Collapse
|
1787
|
Shimazawa R, Ikeda M. Drug–diagnostic co-development: challenges and issues. Expert Rev Mol Diagn 2016; 16:187-204. [DOI: 10.1586/14737159.2016.1132163] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
1788
|
Ajithkumar T, Parkinson C, Fife K, Corrie P, Jefferies S. Evolving treatment options for melanoma brain metastases. Lancet Oncol 2016; 16:e486-97. [PMID: 26433822 DOI: 10.1016/s1470-2045(15)00141-2] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2015] [Revised: 07/04/2015] [Accepted: 07/08/2015] [Indexed: 11/28/2022]
Abstract
Melanoma is a leading cause of lost productivity due to premature cancer mortality. Melanoma frequently spreads to the brain and is associated with rapid deterioration in quality and quantity of life. Until now, treatment options have been restricted to surgery and radiotherapy, although neither modality has been well studied in clinical trials. However, the new immune checkpoint inhibitors and molecularly targeted agents that have been introduced for treatment of metastatic melanoma are active against brain metastases and offer new opportunities to improve disease outcomes. New challenges arise, including how to integrate or sequence multiple treatment modalities, and current practice varies widely. In this Review, we summarise evidence for the treatment of melanoma brain metastases, and discuss the rationale and evidence for combination modalities, highlighting areas for future research.
Collapse
Affiliation(s)
- Thankamma Ajithkumar
- Department of Oncology, Cambridge University Hospitals National Health Service Foundation Trust, Cambridge, UK.
| | - Christine Parkinson
- Department of Oncology, Cambridge University Hospitals National Health Service Foundation Trust, Cambridge, UK
| | - Kate Fife
- Department of Oncology, Cambridge University Hospitals National Health Service Foundation Trust, Cambridge, UK
| | - Pippa Corrie
- Department of Oncology, Cambridge University Hospitals National Health Service Foundation Trust, Cambridge, UK
| | - Sarah Jefferies
- Department of Oncology, Cambridge University Hospitals National Health Service Foundation Trust, Cambridge, UK
| |
Collapse
|
1789
|
Sebaratnam D, Anforth R, Fernández-Peñas P. Treatment-specific utility weightings are needed for cost-utility analysis in metastatic melanoma. Br J Dermatol 2016; 174:462-3. [DOI: 10.1111/bjd.14264] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- D.F. Sebaratnam
- St John's Institute of Dermatology; Guy's and St Thomas' NHS Trust; London U.K
- Department of Dermatology; Westmead Hospital; Westmead NSW 2145 Australia
| | - R. Anforth
- Department of Dermatology; Westmead Hospital; Westmead NSW 2145 Australia
| | - P. Fernández-Peñas
- Department of Dermatology; Westmead Hospital; Westmead NSW 2145 Australia
| |
Collapse
|
1790
|
Xu MM, Pu Y, Zhang Y, Fu YX. The Role of Adaptive Immunity in the Efficacy of Targeted Cancer Therapies. Trends Immunol 2016; 37:141-153. [PMID: 26778079 DOI: 10.1016/j.it.2015.12.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Revised: 12/10/2015] [Accepted: 12/13/2015] [Indexed: 01/22/2023]
Abstract
Accumulating evidence indicates that the efficacy of tumor-targeted therapies relies on the host immune response, including targeted small-molecule and antibody approaches that were not previously thought to have an immune component. Here, we review the current understanding of how targeted therapies on tumor cells could have a major impact on the immune response, and how this relates to the therapeutic efficacy of these approaches. In this context, we evaluate different strategies that combine targeted therapies with immunotherapy approaches, and discuss past and ongoing clinical trials. We highlight gaps in knowledge, and argue that significant progress for combined therapies will require a better understanding of the complex interactions between immune cells, the tumor, and the tumor microenvironment (TME) in different cancer settings.
Collapse
Affiliation(s)
- Meng Michelle Xu
- Department of Pathology and Committee on Immunology, University of Chicago, Chicago, IL 60637, USA
| | - Yang Pu
- Department of Pathology and Committee on Immunology, University of Chicago, Chicago, IL 60637, USA
| | - Yuan Zhang
- Department of Pathology and Committee on Immunology, University of Chicago, Chicago, IL 60637, USA
| | - Yang-Xin Fu
- Department of Pathology and Committee on Immunology, University of Chicago, Chicago, IL 60637, USA; Department of Pathology and Immunology, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA.
| |
Collapse
|
1791
|
Gershenwald JE, Guy GP. Stemming the Rising Incidence of Melanoma: Calling Prevention to Action. J Natl Cancer Inst 2016; 108:djv381. [PMID: 26563358 PMCID: PMC6048594 DOI: 10.1093/jnci/djv381] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2015] [Indexed: 12/16/2022] Open
Affiliation(s)
- Jeffrey E Gershenwald
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Gery P Guy
- Division of Cancer Prevention and Control, Centers for Disease Control and Prevention, Atlanta, Georgia
| |
Collapse
|
1792
|
Zahnreich S, Mayer A, Loquai C, Grabbe S, Schmidberger H. Radiotherapy with BRAF inhibitor therapy for melanoma: progress and possibilities. Future Oncol 2016; 12:95-106. [DOI: 10.2217/fon.15.297] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The introduction of small molecule BRAFV600 kinase inhibitors represents a milestone in the targeted therapy of patients with metastatic melanoma by a significant increase in therapeutic efficacy in terms of overall and progression-free survival compared with conventional chemotherapy. Beside BRAFV600 inhibitor treatment, radiotherapy is a further mainstay for the therapy of metastatic melanoma and thus a concomitant or sequential application of BRAFV600 inhibitors and radiotherapy is inevitable. Recent reports show a significant radiosensitization of the irradiated healthy tissue in patients with melanoma after the combination of radiotherapy and BRAFV600 inhibitors, evoking concern in clinical practice. We review interactions of BRAFV600 inhibitors and radiation with regard to antitumor effects and an increased radiotoxicity in the healthy tissue.
Collapse
Affiliation(s)
- Sebastian Zahnreich
- Department of Radiation Oncology & Radiotherapy, University Medical Center Johannes Gutenberg University Mainz, Germany
| | - Arnulf Mayer
- Department of Radiation Oncology & Radiotherapy, University Medical Center Johannes Gutenberg University Mainz, Germany
| | - Carmen Loquai
- Department of Dermatology, University Medical Center Johannes Gutenberg University Mainz, Germany
| | - Stephan Grabbe
- Department of Dermatology, University Medical Center Johannes Gutenberg University Mainz, Germany
| | - Heinz Schmidberger
- Department of Radiation Oncology & Radiotherapy, University Medical Center Johannes Gutenberg University Mainz, Germany
| |
Collapse
|
1793
|
Abstract
Mucosal melanoma is an exceedingly rare variant of cutaneous melanoma that, due to its rarity, is poorly described and infrequently studied. Primary sites of origin include the head and neck, anorectum and vulvovaginal regions. It is uniquely different from cutaneous melanoma with respect to epidemiology, etiology, pathogenesis and prognosis. The etiology and pathogenesis remain unclear. Unlike cutaneous melanoma, exposure to UV light is not an apparent risk factor. Furthermore, distinct molecular features including a lower incidence of BRAF oncogene mutations but a higher incidence of KIT oncogene mutations suggest divergent genetic etiologies. Mucosal melanomas generally present at a later stage, are more aggressive and carry a worse prognosis regardless of the stage at diagnosis. Establishing standardized treatment guidelines has been challenging due to the rarity of the disease. Early detection provides the best chance at survival but is often difficult due to anatomic location. Surgery remains the primary therapeutic intervention if complete resection is technically feasible given the anatomic location. Radiotherapy may be used to achieve local control when resection is not feasible, or adjuvantly to enhance locoregional control, but most studies have failed to demonstrate an improvement in overall survival. There are no consensus guidelines on the optimal systemic therapy, and regimens are often extrapolated from data based on therapies used to treat advanced cutaneous melanoma. Clinical trials, particularly utilizing newer targeted therapies and immunotherapies, are investigating novel treatment approaches.
Collapse
|
1794
|
Abstract
Next-generation sequencing (NGS) approaches are highly applicable to clinical studies. We review recent advances in sequencing technologies, as well as their benefits and tradeoffs, to provide an overview of clinical genomics from study design to computational analysis. Sequencing technologies enable genomic, transcriptomic, and epigenomic evaluations. Studies that use a combination of whole genome, exome, mRNA, and bisulfite sequencing are now feasible due to decreasing sequencing costs. Single-molecule sequencing increases read length, with the MinIONTM nanopore sequencer, which offers a uniquely portable option at a lower cost. Many of the published comparisons we review here address the challenges associated with different sequencing methods. Overall, NGS techniques, coupled with continually improving analysis algorithms, are useful for clinical studies in many realms, including cancer, chronic illness, and neurobiology. We, and others in the field, anticipate the clinical use of NGS approaches will continue to grow, especially as we shift into an era of precision medicine.
Collapse
Affiliation(s)
- Priyanka Vijay
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, New York. Tri-Institutional Training Program in Computational Biology and Medicine, Weill Cornell Medical College, New York, New York
| | - Alexa B.R. McIntyre
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, New York. Tri-Institutional Training Program in Computational Biology and Medicine, Weill Cornell Medical College, New York, New York
| | - Christopher E. Mason
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, New York. Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medical College, New York, New York. Feil Family Brain and Mind Research Institute, New York, New York
| | - Jeffrey P. Greenfield
- Department of Neurological Surgery, New York-Presbyterian Hospital, Weill Cornell Medical College, New York, New York
| | - Sheng Li
- Department of Neurological Surgery, New York-Presbyterian Hospital, Weill Cornell Medical College, New York, New York
| |
Collapse
|
1795
|
Wu ZL, Fang YL, Tang YT, Xiao MW, Ye J, Li GX, Hu AX. Synthesis and antitumor evaluation of 5-(benzo[d][1,3]dioxol-5-ylmethyl)-4-(tert-butyl)-N-arylthiazol-2-amines. MEDCHEMCOMM 2016. [DOI: 10.1039/c6md00234j] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The strategy for designing target compounds as antitumor agents.
Collapse
Affiliation(s)
- Z. L. Wu
- College of Chemistry and Chemical Engineering
- Hunan University
- Changsha
- China
| | - Y. L. Fang
- College of Chemistry and Chemical Engineering
- Hunan University
- Changsha
- China
| | - Y. T. Tang
- College of Chemistry and Chemical Engineering
- Hunan University
- Changsha
- China
| | - M. W. Xiao
- College of Chemistry and Chemical Engineering
- Hunan University
- Changsha
- China
| | - J. Ye
- College of Chemistry and Chemical Engineering
- Hunan University
- Changsha
- China
| | - G. X. Li
- College of Chemistry and Chemical Engineering
- Hunan University
- Changsha
- China
| | - A. X. Hu
- College of Chemistry and Chemical Engineering
- Hunan University
- Changsha
- China
| |
Collapse
|
1796
|
Yoshida T, Yamaguchi T, Kawasaki H, Sakamoto S. The Use of a Chemical Biology in the Development of a Novel MEK1/2 Inhibitor. J SYN ORG CHEM JPN 2016. [DOI: 10.5059/yukigoseikyokaishi.74.462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Takayuki Yoshida
- Pharmaceutical Frontier Research Laboratories, Central Pharmaceutical Institute, Japan Tobacco
| | - Takayuki Yamaguchi
- Biological/Pharmacological Research Laboratories, Central Pharmaceutical Research Institute, Japan Tobacco
| | - Hisashi Kawasaki
- Chemical Research Laboratories, Central Pharmaceutical Research Institute, Japan Tobacco
| | - Shinji Sakamoto
- Pharmaceutical Frontier Research Laboratories, Central Pharmaceutical Institute, Japan Tobacco
| |
Collapse
|
1797
|
Cytostatic Agents—Tyrosine Kinase Inhibitors Utilized in the Treatment of Solid Malignancies. SIDE EFFECTS OF DRUGS ANNUAL 2016. [DOI: 10.1016/bs.seda.2016.08.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
1798
|
Eroglu Z, Ribas A. Combination therapy with BRAF and MEK inhibitors for melanoma: latest evidence and place in therapy. Ther Adv Med Oncol 2016; 8:48-56. [PMID: 26753005 PMCID: PMC4699264 DOI: 10.1177/1758834015616934] [Citation(s) in RCA: 208] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Treatment with BRAF inhibitors such as vemurafenib or dabrafenib in patients with advanced BRAFV600 mutated melanoma has shown objective tumor responses in approximately half of the patients. However, the duration of responses is limited in a majority of these patients, with progression-free survival rates around 6 months due to tumor progression from development of acquired resistance. Preclinical studies have suggested that concurrent inhibition of the BRAF kinases and MEK of the mitogen-activated protein kinase (MAPK) pathway could decrease MAPK-driven acquired resistance, resulting in longer duration of responses, higher rate of tumor responses, and a decrease in the cutaneous toxicities observed from paradoxical MAPK pathway activation with BRAF inhibitor monotherapy. This review provides an overview of the currently available clinical trial data on BRAF and MEK inhibitors together and in combinations with other therapeutic agents.
Collapse
Affiliation(s)
| | - Antoni Ribas
- Department of Medicine, Division of Hematology-Oncology, UCLA, 11-934 Factor Building, 10833 Le Conte Avenue, Los Angeles, CA 90095-1782, USA
| |
Collapse
|
1799
|
Vilgelm AE, Johnson CA, Prasad N, Yang J, Chen SC, Ayers GD, Pawlikowski JS, Raman D, Sosman JA, Kelley M, Ecsedy JA, Shyr Y, Levy SE, Richmond A. Connecting the Dots: Therapy-Induced Senescence and a Tumor-Suppressive Immune Microenvironment. J Natl Cancer Inst 2015; 108:djv406. [PMID: 26719346 DOI: 10.1093/jnci/djv406] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 12/01/2015] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Tumor cell senescence is a common outcome of anticancer therapy. Here we investigated how therapy-induced senescence (TIS) affects tumor-infiltrating leukocytes (TILs) and the efficacy of immunotherapy in melanoma. METHODS Tumor senescence was induced by AURKA or CDK4/6 inhibitors (AURKAi, CDK4/6i). Transcriptomes of six mouse tumors with differential response to AURKAi were analyzed by RNA sequencing, and TILs were characterized by flow cytometry. Chemokine RNA and protein expression were determined by quantitative real-time polymerase chain reaction and enzyme-linked immunosorbent assay. Therapeutic response was queried in immunodeficient mice, in mice with CCL5-deficient tumors, and in mice cotreated with CD137 agonist to activate TILs. CCL5 expression in reference to TIS and markers of TILs was studied in human melanoma tumors using patient-derived xenografts (n = 3 patients, n = 3 mice each), in AURKAi clinical trial samples (n = 3 patients, before/after therapy), and in The Cancer Genome Atlas (n = 278). All statistical tests were two-sided. RESULTS AURKAi response was associated with induction of the immune transcriptome (P = 3.5 x 10-29) while resistance inversely correlated with TIL numbers (Spearman r = -0.87, P < .001). AURKAi and CDK4/6i promoted the recruitment of TILs by inducing CCL5 secretion in melanoma cells (P ≤ .005) in an NF-κB-dependent manner. Therapeutic response to AURKAi was impaired in immunodeficient compared with immunocompetent mice (0% vs 67% tumors regressed, P = .01) and in mice bearing CCL5-deficient vs control tumors (P = .61 vs P = .02); however, AURKAi response was greatly enhanced in mice also receiving T-cell-activating immunotherapy (P < .001). In human tumors, CCL5 expression was also induced by AURKAi (P ≤ .02) and CDK4/6i (P = .01) and was associated with increased immune marker expression (P = 1.40 x 10-93). CONCLUSIONS Senescent melanoma cells secret CCL5, which promotes recruitment of TILs. Combining TIS with immunotherapy that enhances tumor cell killing by TILs is a promising novel approach to improve melanoma outcomes.
Collapse
Affiliation(s)
- Anna E Vilgelm
- Tennessee Valley Healthcare System, Department of Veterans Affairs, Nashville, TN (AEV, CAJ, JY, JSP, DR, AR); Department of Cancer Biology (AEV, AJ, JY, SCC, JSP, DR, AR), Center for Quantitative Sciences (SCC), Division of Cancer Biostatistics, Department of Biostatistics (GDA, YS), Division of Hematology/Oncology, Department of Medicine (JAS), Vanderbilt University Medical Center, Nashville, TN; HudsonAlpha Institute for Biotechnology, Huntsville, Alabama (NP, SEL); Division of Surgical Oncology, Department of Surgery, Vanderbilt University School of Medicine, Nashville, TN (MK); Translational Medicine, Takeda Pharmaceuticals International Co, Cambridge, MA (JAE).
| | - C Andrew Johnson
- Tennessee Valley Healthcare System, Department of Veterans Affairs, Nashville, TN (AEV, CAJ, JY, JSP, DR, AR); Department of Cancer Biology (AEV, AJ, JY, SCC, JSP, DR, AR), Center for Quantitative Sciences (SCC), Division of Cancer Biostatistics, Department of Biostatistics (GDA, YS), Division of Hematology/Oncology, Department of Medicine (JAS), Vanderbilt University Medical Center, Nashville, TN; HudsonAlpha Institute for Biotechnology, Huntsville, Alabama (NP, SEL); Division of Surgical Oncology, Department of Surgery, Vanderbilt University School of Medicine, Nashville, TN (MK); Translational Medicine, Takeda Pharmaceuticals International Co, Cambridge, MA (JAE)
| | - Nripesh Prasad
- Tennessee Valley Healthcare System, Department of Veterans Affairs, Nashville, TN (AEV, CAJ, JY, JSP, DR, AR); Department of Cancer Biology (AEV, AJ, JY, SCC, JSP, DR, AR), Center for Quantitative Sciences (SCC), Division of Cancer Biostatistics, Department of Biostatistics (GDA, YS), Division of Hematology/Oncology, Department of Medicine (JAS), Vanderbilt University Medical Center, Nashville, TN; HudsonAlpha Institute for Biotechnology, Huntsville, Alabama (NP, SEL); Division of Surgical Oncology, Department of Surgery, Vanderbilt University School of Medicine, Nashville, TN (MK); Translational Medicine, Takeda Pharmaceuticals International Co, Cambridge, MA (JAE)
| | - Jinming Yang
- Tennessee Valley Healthcare System, Department of Veterans Affairs, Nashville, TN (AEV, CAJ, JY, JSP, DR, AR); Department of Cancer Biology (AEV, AJ, JY, SCC, JSP, DR, AR), Center for Quantitative Sciences (SCC), Division of Cancer Biostatistics, Department of Biostatistics (GDA, YS), Division of Hematology/Oncology, Department of Medicine (JAS), Vanderbilt University Medical Center, Nashville, TN; HudsonAlpha Institute for Biotechnology, Huntsville, Alabama (NP, SEL); Division of Surgical Oncology, Department of Surgery, Vanderbilt University School of Medicine, Nashville, TN (MK); Translational Medicine, Takeda Pharmaceuticals International Co, Cambridge, MA (JAE)
| | - Sheau-Chiann Chen
- Tennessee Valley Healthcare System, Department of Veterans Affairs, Nashville, TN (AEV, CAJ, JY, JSP, DR, AR); Department of Cancer Biology (AEV, AJ, JY, SCC, JSP, DR, AR), Center for Quantitative Sciences (SCC), Division of Cancer Biostatistics, Department of Biostatistics (GDA, YS), Division of Hematology/Oncology, Department of Medicine (JAS), Vanderbilt University Medical Center, Nashville, TN; HudsonAlpha Institute for Biotechnology, Huntsville, Alabama (NP, SEL); Division of Surgical Oncology, Department of Surgery, Vanderbilt University School of Medicine, Nashville, TN (MK); Translational Medicine, Takeda Pharmaceuticals International Co, Cambridge, MA (JAE)
| | - Gregory D Ayers
- Tennessee Valley Healthcare System, Department of Veterans Affairs, Nashville, TN (AEV, CAJ, JY, JSP, DR, AR); Department of Cancer Biology (AEV, AJ, JY, SCC, JSP, DR, AR), Center for Quantitative Sciences (SCC), Division of Cancer Biostatistics, Department of Biostatistics (GDA, YS), Division of Hematology/Oncology, Department of Medicine (JAS), Vanderbilt University Medical Center, Nashville, TN; HudsonAlpha Institute for Biotechnology, Huntsville, Alabama (NP, SEL); Division of Surgical Oncology, Department of Surgery, Vanderbilt University School of Medicine, Nashville, TN (MK); Translational Medicine, Takeda Pharmaceuticals International Co, Cambridge, MA (JAE)
| | - Jeff S Pawlikowski
- Tennessee Valley Healthcare System, Department of Veterans Affairs, Nashville, TN (AEV, CAJ, JY, JSP, DR, AR); Department of Cancer Biology (AEV, AJ, JY, SCC, JSP, DR, AR), Center for Quantitative Sciences (SCC), Division of Cancer Biostatistics, Department of Biostatistics (GDA, YS), Division of Hematology/Oncology, Department of Medicine (JAS), Vanderbilt University Medical Center, Nashville, TN; HudsonAlpha Institute for Biotechnology, Huntsville, Alabama (NP, SEL); Division of Surgical Oncology, Department of Surgery, Vanderbilt University School of Medicine, Nashville, TN (MK); Translational Medicine, Takeda Pharmaceuticals International Co, Cambridge, MA (JAE)
| | - Dayanidhi Raman
- Tennessee Valley Healthcare System, Department of Veterans Affairs, Nashville, TN (AEV, CAJ, JY, JSP, DR, AR); Department of Cancer Biology (AEV, AJ, JY, SCC, JSP, DR, AR), Center for Quantitative Sciences (SCC), Division of Cancer Biostatistics, Department of Biostatistics (GDA, YS), Division of Hematology/Oncology, Department of Medicine (JAS), Vanderbilt University Medical Center, Nashville, TN; HudsonAlpha Institute for Biotechnology, Huntsville, Alabama (NP, SEL); Division of Surgical Oncology, Department of Surgery, Vanderbilt University School of Medicine, Nashville, TN (MK); Translational Medicine, Takeda Pharmaceuticals International Co, Cambridge, MA (JAE)
| | - Jeffrey A Sosman
- Tennessee Valley Healthcare System, Department of Veterans Affairs, Nashville, TN (AEV, CAJ, JY, JSP, DR, AR); Department of Cancer Biology (AEV, AJ, JY, SCC, JSP, DR, AR), Center for Quantitative Sciences (SCC), Division of Cancer Biostatistics, Department of Biostatistics (GDA, YS), Division of Hematology/Oncology, Department of Medicine (JAS), Vanderbilt University Medical Center, Nashville, TN; HudsonAlpha Institute for Biotechnology, Huntsville, Alabama (NP, SEL); Division of Surgical Oncology, Department of Surgery, Vanderbilt University School of Medicine, Nashville, TN (MK); Translational Medicine, Takeda Pharmaceuticals International Co, Cambridge, MA (JAE)
| | - Mark Kelley
- Tennessee Valley Healthcare System, Department of Veterans Affairs, Nashville, TN (AEV, CAJ, JY, JSP, DR, AR); Department of Cancer Biology (AEV, AJ, JY, SCC, JSP, DR, AR), Center for Quantitative Sciences (SCC), Division of Cancer Biostatistics, Department of Biostatistics (GDA, YS), Division of Hematology/Oncology, Department of Medicine (JAS), Vanderbilt University Medical Center, Nashville, TN; HudsonAlpha Institute for Biotechnology, Huntsville, Alabama (NP, SEL); Division of Surgical Oncology, Department of Surgery, Vanderbilt University School of Medicine, Nashville, TN (MK); Translational Medicine, Takeda Pharmaceuticals International Co, Cambridge, MA (JAE)
| | - Jeffrey A Ecsedy
- Tennessee Valley Healthcare System, Department of Veterans Affairs, Nashville, TN (AEV, CAJ, JY, JSP, DR, AR); Department of Cancer Biology (AEV, AJ, JY, SCC, JSP, DR, AR), Center for Quantitative Sciences (SCC), Division of Cancer Biostatistics, Department of Biostatistics (GDA, YS), Division of Hematology/Oncology, Department of Medicine (JAS), Vanderbilt University Medical Center, Nashville, TN; HudsonAlpha Institute for Biotechnology, Huntsville, Alabama (NP, SEL); Division of Surgical Oncology, Department of Surgery, Vanderbilt University School of Medicine, Nashville, TN (MK); Translational Medicine, Takeda Pharmaceuticals International Co, Cambridge, MA (JAE)
| | - Yu Shyr
- Tennessee Valley Healthcare System, Department of Veterans Affairs, Nashville, TN (AEV, CAJ, JY, JSP, DR, AR); Department of Cancer Biology (AEV, AJ, JY, SCC, JSP, DR, AR), Center for Quantitative Sciences (SCC), Division of Cancer Biostatistics, Department of Biostatistics (GDA, YS), Division of Hematology/Oncology, Department of Medicine (JAS), Vanderbilt University Medical Center, Nashville, TN; HudsonAlpha Institute for Biotechnology, Huntsville, Alabama (NP, SEL); Division of Surgical Oncology, Department of Surgery, Vanderbilt University School of Medicine, Nashville, TN (MK); Translational Medicine, Takeda Pharmaceuticals International Co, Cambridge, MA (JAE)
| | - Shawn E Levy
- Tennessee Valley Healthcare System, Department of Veterans Affairs, Nashville, TN (AEV, CAJ, JY, JSP, DR, AR); Department of Cancer Biology (AEV, AJ, JY, SCC, JSP, DR, AR), Center for Quantitative Sciences (SCC), Division of Cancer Biostatistics, Department of Biostatistics (GDA, YS), Division of Hematology/Oncology, Department of Medicine (JAS), Vanderbilt University Medical Center, Nashville, TN; HudsonAlpha Institute for Biotechnology, Huntsville, Alabama (NP, SEL); Division of Surgical Oncology, Department of Surgery, Vanderbilt University School of Medicine, Nashville, TN (MK); Translational Medicine, Takeda Pharmaceuticals International Co, Cambridge, MA (JAE)
| | - Ann Richmond
- Tennessee Valley Healthcare System, Department of Veterans Affairs, Nashville, TN (AEV, CAJ, JY, JSP, DR, AR); Department of Cancer Biology (AEV, AJ, JY, SCC, JSP, DR, AR), Center for Quantitative Sciences (SCC), Division of Cancer Biostatistics, Department of Biostatistics (GDA, YS), Division of Hematology/Oncology, Department of Medicine (JAS), Vanderbilt University Medical Center, Nashville, TN; HudsonAlpha Institute for Biotechnology, Huntsville, Alabama (NP, SEL); Division of Surgical Oncology, Department of Surgery, Vanderbilt University School of Medicine, Nashville, TN (MK); Translational Medicine, Takeda Pharmaceuticals International Co, Cambridge, MA (JAE)
| |
Collapse
|
1800
|
Mitsis D, Opyrchal M, Zhao Y, Kane Iii JM, Cheney R, Salerno KE. Exceptional Clinical Response to BRAF-Targeted Therapy in a Patient with Metastatic Sarcoma. Cureus 2015; 7:e439. [PMID: 26858920 PMCID: PMC4731329 DOI: 10.7759/cureus.439] [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: 11/23/2015] [Accepted: 12/28/2015] [Indexed: 11/27/2022] Open
Abstract
Soft tissue sarcomas (STS) are a rare and heterogeneous group of tumors arising from mesenchymal tissue comprising 1% of all adult cancers. The prognosis of metastatic STS is dismal. As there are few active drugs, there is a critical need to find new therapeutic alternatives in order to improve outcomes. Most sarcoma subtypes are fairly resistant to standard chemotherapy regimens and/or have a short duration of response. The era of molecular targeted therapy may present new treatment options for metastatic STS and an opportunity to drive biomarker discovery and personalized medicine. This case report describes a patient with a synchronous metastatic high-grade sarcoma who was treated with BRAF-targeted therapy with resolution of his metastatic lesions. To our knowledge, this exceptional response has not been described in sarcoma literature. The patient presented with a large anterior abdominopelvic wall mass. Biopsy showed a high-grade spindle cell sarcoma. Staging scans confirmed two pulmonary metastases. He was initiated on combination chemotherapy with mixed results. He received 50 Gy of radiation to the primary tumor followed by two additional cycles of combination chemotherapy, with an interval increase in the size of the pulmonary metastases. Molecular testing revealed a BRAF V600E mutation in the primary tumor. The patient was initiated on dabrafenib/trametinib with a dramatic response and resolution of his pulmonary metastases. The patient underwent surgical resection of his primary mass, and pathology confirmed no evidence of residual disease. Detailed genetic characterization of STS coupled with novel therapeutic strategies, including molecular targeted therapy, may have the potential to transform the care of patients diagnosed with sarcoma.
Collapse
Affiliation(s)
| | | | - Yujie Zhao
- Medical Oncology, Carle Foundation Hospital
| | | | | | | |
Collapse
|