951
|
Stadler R, Leiter U, Garbe C. Lack of survival benefit in sentinel lymph node-positive melanoma with immediate complete lymphadenectomy - a review. J Dtsch Dermatol Ges 2018; 17:7-13. [DOI: 10.1111/ddg.13707] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 09/11/2018] [Indexed: 11/30/2022]
Affiliation(s)
- Rudolf Stadler
- University Hospital for Dermatology; Johannes Wesling Clinical Centre in Minden; Ruhr University Hospital; Bochum Germany
| | - Ulrike Leiter
- Centre for Dermato-oncology; University Department of Dermatology, Eberhard Karls University; Tübingen Germany
| | - Claus Garbe
- Centre for Dermato-oncology; University Department of Dermatology, Eberhard Karls University; Tübingen Germany
| |
Collapse
|
952
|
Gu F, Chen TH, Pfeiffer RM, Fargnoli MC, Calista D, Ghiorzo P, Peris K, Puig S, Menin C, De Nicolo A, Rodolfo M, Pellegrini C, Pastorino L, Evangelou E, Zhang T, Hua X, DellaValle CT, Timothy Bishop D, MacGregor S, Iles MI, Law MH, Cust A, Brown KM, Stratigos AJ, Nagore E, Chanock S, Shi J, Consortium MMA, Consortium M, Landi MT. Combining common genetic variants and non-genetic risk factors to predict risk of cutaneous melanoma. Hum Mol Genet 2018; 27:4145-4156. [PMID: 30060076 PMCID: PMC6240742 DOI: 10.1093/hmg/ddy282] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 06/14/2018] [Accepted: 07/24/2018] [Indexed: 02/04/2023] Open
Abstract
Melanoma heritability is among the highest for cancer and single nucleotide polymorphisms (SNPs) contribute to it. To date, only SNPs that reached statistical significance in genome-wide association studies or few candidate SNPs have been included in melanoma risk prediction models. We compared four approaches for building polygenic risk scores (PRS) using 12 874 melanoma cases and 23 203 controls from Melanoma Meta-Analysis Consortium as a training set, and newly genotyped 3102 cases and 2301 controls from the MelaNostrum consortium for validation. We estimated adjusted odds ratios (ORs) for melanoma risk using traditional melanoma risk factors and the PRS with the largest area under the receiver operator characteristics curve (AUC). We estimated absolute risks combining the PRS and other risk factors, with age- and sex-specific melanoma incidence and competing mortality rates from Italy as an example. The best PRS, including 204 SNPs (AUC = 64.4%; 95% confidence interval (CI) = 63-65.8%), developed using winner's curse estimate corrections, had a per-quintile OR = 1.35 (95% CI = 1.30-1.41), corresponding to a 3.33-fold increase comparing the 5th to the 1st PRS quintile. The AUC improvement by adding the PRS was up to 7%, depending on adjusted factors and country. The 20-year absolute risk estimates based on the PRS, nevus count and pigmentation characteristics for a 60-year-old Italian man ranged from 0.5 to 11.8% (relative risk = 26.34), indicating good separation.
Collapse
Affiliation(s)
- Fangyi Gu
- Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ting-Huei Chen
- Department of Mathematics and Statistics, Laval University, Quebec, Canada
| | - Ruth M Pfeiffer
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | | | - Donato Calista
- Department of Dermatology, Maurizio Bufalini Hospital, Cesena, Italy
| | - Paola Ghiorzo
- Department of Internal Medicine and Medical Specialties, University of Genoa and Genetics of Rare Cancers, Ospedale Policlinico San Martino, Genoa, Italy
| | - Ketty Peris
- Institute of Dermatology, Catholic University, Rome, Italy
| | - Susana Puig
- Dermatology Department, Melanoma Unit, Hospital Clínic de Barcelona, IDIBAPS, Universitat de Barcelona, Barcelona, Spain and Centro de Investigación Biomédica en Red en Enfermedades Raras (CIBERER), Valencia, Spain
| | - Chiara Menin
- Department of Immunology and Molecular Oncology, Veneto Institute of Oncology IOV–IRCCS, Padua, Italy
| | - Arcangela De Nicolo
- Cancer Genomics Program, Veneto Institute of Oncology IOV–IRCCS, Padua, Italy
| | - Monica Rodolfo
- Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | | | - Lorenza Pastorino
- Department of Internal Medicine and Medical Specialties, University of Genoa and Genetics of Rare Cancers, Ospedale Policlinico San Martino, Genoa, Italy
| | - Evangelos Evangelou
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
| | - Tongwu Zhang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Xing Hua
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Curt T DellaValle
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - D Timothy Bishop
- Section of Epidemiology and Biostatistics, Leeds Institute of Cancer and Pathology, University of Leeds, UK
| | - Stuart MacGregor
- Statistical Genetics, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Mark I Iles
- Section of Epidemiology and Biostatistics, Leeds Institute of Cancer and Pathology, University of Leeds, UK
| | - Matthew H Law
- Statistical Genetics, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Anne Cust
- Sydney School of Public Health, and Melanoma Institute Australia, The University of Sydney, Sydney, Australia
| | - Kevin M Brown
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Alexander J Stratigos
- 1 Department of Dermatology–Venereology, National and Kapodistrian University of Athens School of Medicine, Andreas Sygros Hospital, Athens, Greece
| | - Eduardo Nagore
- Department of Dermatology, Instituto Valenciano de Oncología, València, Spain
| | - Stephen Chanock
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jianxin Shi
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | | | | | - Maria Teresa Landi
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
953
|
Abstract
INTRODUCTION Protein kinases are involved in various cellular functions. About 2% of the human genome encodes for protein kinases. Dysregulation of protein kinases is implicated in various processes of carcinogenesis. The advent of protein kinase inhibitors in cancer therapy has led to a paradigm shift in cancer therapy. Several protein kinase inhibitors have been approved by FDA in the last few decades. Areas covered: This article provides a review of the FDA approved protein kinase inhibitors as of December 2017 for the well-known oncogenic protein kinases. A list of FDA approved protein kinase inhibitors and their FDA approved clinical indications were cataloged. The role of the respective oncogenic protein kinases in carcinogenesis and cancer progression and the relevant landmark clinical trials of respective protein kinase inhibitors leading up to the FDA approval were PubMed searched and discussed. Expert commentary: Further understanding of the molecular origin of various cancers would help identify new targets. Use of biomarker profiling might select the patient population that would benefit better from kinase inhibitors. Clinical trials should be designed to identify the appropriate sequence of the available kinase inhibitors. It would prove to be useful to test these drugs in the adjuvant setting.
Collapse
Affiliation(s)
- Radhamani Kannaiyan
- University of Arizona College of Medicine at South Campus, Tucson, Arizona, USA
| | | |
Collapse
|
954
|
Grob JJ, Garbe C, Ascierto P, Larkin J, Dummer R, Schadendorf D. Adjuvant melanoma therapy with new drugs: should physicians continue to focus on metastatic disease or use it earlier in primary melanoma? Lancet Oncol 2018; 19:e720-e725. [DOI: 10.1016/s1470-2045(18)30596-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 08/01/2018] [Accepted: 08/02/2018] [Indexed: 02/06/2023]
|
955
|
Abstract
Cardio-oncology has emerged as best option for many patients with cardiovascular complications related to cancer and cancer therapy. Classical chemotherapy, targeted and immune therapies as wells as radiotherapy challenge the cardiovascular system at multiple levels, including increased rates of e.g., hypertension, coronary artery disease, cardiomyopathy, arrhythmia, and thrombosis. The cardiologist working within the cardio-oncology team is confronted with a broad spectrum of therapies and combination protocols. Evidence from the past few years implicate that at least 30 novel cancer drugs can be expected to receive approval each year. The rate and extent of cardiovascular complications particularly for these new therapies remains to be evaluated. Overall, cardio-oncology has several important tasks to establish an optimal care for cancer patients: (I) risk assessment before therapy to prevent onset of cardiovascular disease, (II) assessment of diagnostic pathways to provide a timely diagnosis of early stages of cardiovascular complications, (III) characterize the extent of morbidity and mortality as induced by cancer therapies, (IV) establish therapeutic options to treat cardiotoxicity and (V) monitor long-term cancer survivors. In this special edition, experts in the field provide an overview about key concepts in cardio-oncology. This is complemented by general considerations about novel concepts in cardio-oncology including the establishment of fellowship programs, ethical issue and cancer survivorship programs as outlined in the present review.
Collapse
Affiliation(s)
- Tienush Rassaf
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, Medical Faculty, University Hospital Essen, 45147 Essen, Germany
| | - Matthias Totzeck
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, Medical Faculty, University Hospital Essen, 45147 Essen, Germany
| |
Collapse
|
956
|
El Rassy E, Farhat F, Kattan J. The forgotten role of adjuvant immune checkpoint inhibitors in preventing melanoma brain metastasis. Immunotherapy 2018; 10:1289-1291. [PMID: 30474477 DOI: 10.2217/imt-2018-0108] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
- Elie El Rassy
- Department of Medical Oncology, Hotel Dieu de France University Hospital, Faculty of Medicine, Saint Joseph University, Lebanon
| | - Fadi Farhat
- Department of Medical Oncology, Hotel Dieu de France University Hospital, Faculty of Medicine, Saint Joseph University, Lebanon
| | - Joseph Kattan
- Department of Medical Oncology, Hotel Dieu de France University Hospital, Faculty of Medicine, Saint Joseph University, Lebanon
| |
Collapse
|
957
|
Health-related quality of life analysis in stage III melanoma patients treated with adjuvant dendritic cell therapy. Clin Transl Oncol 2018; 21:774-780. [PMID: 30465182 DOI: 10.1007/s12094-018-1987-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 11/09/2018] [Indexed: 10/27/2022]
Abstract
BACKGROUND Health-related quality of life (HRQoL) is an important issue in the rapidly evolving field of adjuvant treatment for stage III melanoma. Dendritic cell vaccination is one of the adjuvant forms of therapy currently investigated. METHODS We enrolled adults with stage III melanoma to receive adjuvant dendritic cell vaccination after a complete radical lymph node dissection. HRQoL assessment was one of the secondary endpoints of this trial and investigated with the EORTC-QLQ-C30 questionnaire at baseline and week 26. RESULTS Fifteen patients with a median age of 50 years were included in the study, with twelve evaluable patients on study at time of the second questionnaire. Global health status and role functioning improved clinically relevant with a mean difference of 15 (p = 0.010) and 26 points (p = 0.005), respectively. DISCUSSION Despite the small number of patients, we found a clinically relevant improved global health status. Besides, compared to the other investigated therapies, toxicity of dendritic cell vaccination is low, which supports our finding. CONCLUSION This is the first description of HRQoL in melanoma patients receiving dendritic cell vaccination. We show the expected improvement in global health status after surgical treatment of stage III melanoma. Thus, adjuvant dendritic cell vaccination does not seem to hamper this improvement, as shown in our small explorative study.
Collapse
|
958
|
Bertolli E, Franke V, Calsavara VF, de Macedo MP, Pinto CAL, van Houdt WJ, Wouters MWJM, Duprat Neto JP, van Akkooi ACJ. Validation of a Nomogram for Non-sentinel Node Positivity in Melanoma Patients, and Its Clinical Implications: A Brazilian–Dutch Study. Ann Surg Oncol 2018; 26:395-405. [DOI: 10.1245/s10434-018-7038-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Indexed: 12/14/2022]
|
959
|
Falzone L, Salomone S, Libra M. Evolution of Cancer Pharmacological Treatments at the Turn of the Third Millennium. Front Pharmacol 2018; 9:1300. [PMID: 30483135 PMCID: PMC6243123 DOI: 10.3389/fphar.2018.01300] [Citation(s) in RCA: 546] [Impact Index Per Article: 78.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 10/23/2018] [Indexed: 12/11/2022] Open
Abstract
The medical history of cancer began millennia ago. Historical findings of patients with cancer date back to ancient Egyptian and Greek civilizations, where this disease was predominantly treated with radical surgery and cautery that were often ineffective, leading to the death of patients. Over the centuries, important discoveries allowed to identify the biological and pathological features of tumors, without however contributing to the development of effective therapeutic approaches until the end of the 1800s, when the discovery of X-rays and their use for the treatment of tumors provided the first modern therapeutic approach in medical oncology. However, a real breakthrough took place after the Second World War, with the discovery of cytotoxic antitumor drugs and the birth of chemotherapy for the treatment of various hematological and solid tumors. Starting from this epochal turning point, there has been an exponential growth of studies concerning the use of new drugs for cancer treatment. The second fundamental breakthrough in the field of oncology and pharmacology took place at the beginning of the '80s, thanks to molecular and cellular biology studies that allowed the development of specific drugs for some molecular targets involved in neoplastic processes, giving rise to targeted therapy. Both chemotherapy and target therapy have significantly improved the survival and quality of life of cancer patients inducing sometimes complete tumor remission. Subsequently, at the turn of the third millennium, thanks to genetic engineering studies, there was a further advancement of clinical oncology and pharmacology with the introduction of monoclonal antibodies and immune checkpoint inhibitors for the treatment of advanced or metastatic tumors, for which no effective treatment was available before. Today, cancer research is always aimed at the study and development of new therapeutic approaches for cancer treatment. Currently, several researchers are focused on the development of cell therapies, anti-tumor vaccines, and new biotechnological drugs that have already shown promising results in preclinical studies, therefore, in the near future, we will certainly assist to a new revolution in the field of medical oncology.
Collapse
Affiliation(s)
- Luca Falzone
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Salvatore Salomone
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy.,Research Center for Prevention, Diagnosis and Treatment of Cancer (PreDiCT), University of Catania, Catania, Italy
| | - Massimo Libra
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy.,Research Center for Prevention, Diagnosis and Treatment of Cancer (PreDiCT), University of Catania, Catania, Italy
| |
Collapse
|
960
|
Nan Tie E, Henderson MA, Gyorki DE. Management of in-transit melanoma metastases: a review. ANZ J Surg 2018; 89:647-652. [PMID: 30414233 DOI: 10.1111/ans.14921] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2018] [Revised: 09/04/2018] [Accepted: 09/25/2018] [Indexed: 02/06/2023]
Abstract
In-transit metastases (ITM) of cutaneous melanoma are locoregional recurrences confined to the superficial lymphatics that occur in 3.4-6.2% of patients diagnosed with melanoma. ITM are a heterogeneous disease that poses a therapeutic dilemma. Patients may have a prolonged disease trajectory involving multiple or repeat treatment modalities for frequent recurrences. The management of ITM has evolved without the development of a standardized protocol. Owing to the variability of the disease course there are few dedicated clinical trials, with a number of key trials in stage III melanoma excluding ITM patients. Thus, there is a paucity of quality data on the efficacy of the treatment modalities available for ITM and even fewer studies directly comparing modalities. At present the mainstay of ITM treatment is surgical resection, with intralesional therapies, isolated limb infusion and radiotherapy utilized as second-line measures. The developing role of targeted therapies and immunotherapy has yet to be explored completely in these patients. This review addresses the evidence base of the efficacy of the various treatment modalities available and those factors that have impacted their clinical uptake.
Collapse
Affiliation(s)
- Emilia Nan Tie
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Michael A Henderson
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Department of Surgery, St Vincent's Hospital, The University of Melbourne, Melbourne, Victoria, Australia
| | - David E Gyorki
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Department of Surgery, St Vincent's Hospital, The University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
961
|
Is earlier better for melanoma checkpoint blockade? Nat Med 2018; 24:1645-1648. [PMID: 30401867 DOI: 10.1038/s41591-018-0250-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 10/09/2018] [Indexed: 11/08/2022]
Abstract
A neoadjuvant approach relying on the administration of combined anti-CTLA-4-anti-PD-1 treatment before lymph node surgery is evaluated in two phase 1 trials. Encouraging clinical, pathological and immunological responses to neoadjuvant therapy were observed, suggesting that this concept warrants further exploration; however, any future approach must address the unacceptably high toxicity of the regimens evaluated in these trials.
Collapse
|
962
|
Bertolli E, de Macedo MP, Calsavara VF, Pinto CAL, Duprat Neto JP. A nomogram to identify high-risk melanoma patients with a negative sentinel lymph node biopsy. J Am Acad Dermatol 2018; 80:722-726. [PMID: 30395920 DOI: 10.1016/j.jaad.2018.10.060] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Revised: 10/16/2018] [Accepted: 10/28/2018] [Indexed: 12/23/2022]
Abstract
BACKGROUND Melanoma patients with negative nodes after sentinel lymph node biopsy are a heterogeneous group. Current guidelines fail to adequately stratify surveillance and treatment for this group. Also, there is scarce data on adjuvant treatments for these patients. OBJECTIVE To create a nomogram including clinical and pathologic characteristics capable of evaluating the risk for recurrence of primary melanoma patients with negative sentinel lymph node biopsies (SLNBs). METHODS We used a retrospective cohort of patients who underwent SLNB during 2000-2015 at a single institution. RESULTS Our cohort comprised 1213 patients. Among these patients, 967 (79.7%) had a negative SLNB, and mean follow-up was 59.67 months. There were 133 recurrences (13.8%); 45 (33.8%) presented with nodal recurrence, and 35 (26.3%) recurred where a SLNB was performed. Breslow thickness, ulceration, and microsatellitosis were found to be predictive of risk for recurrence at 1, 2, 5, and 10 years. LIMITATION Single center analysis. CONCLUSION We created a predictive nomogram for melanoma patients with negative SLNBs. This nomogram is easy to use and identifies high-risk patients who should have more strict surveillance and be considered for adjuvant treatment.
Collapse
Affiliation(s)
- Eduardo Bertolli
- Skin Cancer Department, AC Camargo Cancer Center, São Paulo, Brazil.
| | | | | | | | | |
Collapse
|
963
|
Pampena MB, Cartar HC, Cueto GR, Levy EM, Blanco PA, Barrio MM, Mordoh J. Dissecting the Immune Stimulation Promoted by CSF-470 Vaccine Plus Adjuvants in Cutaneous Melanoma Patients: Long Term Antitumor Immunity and Short Term Release of Acute Inflammatory Reactants. Front Immunol 2018; 9:2531. [PMID: 30450100 PMCID: PMC6224428 DOI: 10.3389/fimmu.2018.02531] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 10/15/2018] [Indexed: 12/21/2022] Open
Abstract
As cutaneous melanoma (CM) currently remains with a bleak prognosis, thorough investigation of new treatment options are of utmost relevance. In the phase II/III randomized clinical trial (CASVAC-0401), the repeated immunization of stages IIB-III CM patients with the irradiated, allogeneic cellular CSF-470 vaccine plus the adjuvants bacillus Calmette-Guerin (BCG) and recombinant human granulocyte macrophage colony-stimulating factor (rhGM-CSF) demonstrated a significant benefit over IFN-alpha2B treatment in distant metastasis-free survival. Here we present on the short and long term immune monitoring results after completing the 2-year protocol; a continuation of the previous report by Mordoh et al. (1). We demonstrate that the repeated CSF-470 vaccinations stimulated a long term cellular and humoral immunity response directed against the vaccine antigens. In the case of 2 patients, we are able to show that a similar immune response was generated against autologous antigens. Evaluation of inhibitory receptor co-expression on patient's T cells indicates that the vaccination protocol did not stimulate T cell exhaustion. In order to better understand the basis for the efficacious vaccine responses observed, we investigated the short term immune events following vaccine injection. A significant increase in C-reactive protein (CRP) and IL-6 was observed 24 h after vaccination, with in vitro studies suggesting IL-6 production occurs in the vaccine site. We demonstrate that CRP enhances the cytotoxicity of peripheral blood mononuclear cells (PBMC) against melanoma cells in an in vitro model. Additionally, CRP stimulates the release of pro and anti-inflammatory cytokines from PBMC. As our results demonstrate that successive vaccinations with CSF-470 plus adjuvants promoted an increase in both anti-tumor innate and adaptive immunity, we propose a subsequent model of action.
Collapse
Affiliation(s)
- María B Pampena
- Centro de Investigaciones Oncológicas-Fundación Cáncer, Buenos Aires, Argentina
| | - Holliday C Cartar
- Centro de Investigaciones Oncológicas-Fundación Cáncer, Buenos Aires, Argentina
| | - Gerardo Rubén Cueto
- Grupo de Bioestadística Aplicada, Departamento de Ecología, Genética y Evolución, Instituto de Ecología, Genética y Evolución de Buenos Aires (IEGEBA-UBA/CONICET), Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Estrella M Levy
- Centro de Investigaciones Oncológicas-Fundación Cáncer, Buenos Aires, Argentina
| | - Paula A Blanco
- Centro de Investigaciones Oncológicas-Fundación Cáncer, Buenos Aires, Argentina
| | - María M Barrio
- Centro de Investigaciones Oncológicas-Fundación Cáncer, Buenos Aires, Argentina
| | - José Mordoh
- Centro de Investigaciones Oncológicas-Fundación Cáncer, Buenos Aires, Argentina.,Department of Biotherapy, Instituto Alexander Fleming, Buenos Aires, Argentina.,Fundación Instituto Leloir, IIBBA-CONICET, Buenos Aires, Argentina
| |
Collapse
|
964
|
Kudchadkar RR, Michielin O, van Akkooi ACJ. Practice-Changing Developments in Stage III Melanoma: Surgery, Adjuvant Targeted Therapy, and Immunotherapy. Am Soc Clin Oncol Educ Book 2018; 38:759-762. [PMID: 30231370 DOI: 10.1200/edbk_200241] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
In this article, we will focus on the practice-changing developments for stage III melanoma, from the use of the sentinel node (SN) biopsy to complete lymph node dissection (CLND) and upcoming adjuvant therapies. MSLT-1 (Multicenter Selective Lymphadenectomy Trial-1) was the first and only prospective randomized controlled trial to examine whether the SN biopsy has any notable melanoma-specific survival benefit (primary endpoint). MSLT-1 randomly assigned 2,001 patients to undergo either wide local excision (WLE) and an SN biopsy or WLE and nodal observation. Two prospective randomized controlled trials have examined the potential benefit for immediate CLND versus delayed CLND after sequential observation. Both the DECOG-SLT and MSLT-2 trials failed to demonstrate a notable benefit for immediate CLND; therefore, sequential follow-up with ultrasonography and a delayed CLND in the case of relapse should be considered the new standard of care. The CheckMate 238 study demonstrated a notable benefit for adjuvant nivolumab in terms of 18-month relapse-free survival (RFS) rates compared with high-dose adjuvant ipilimumab. Single-agent adjuvant BRAF inhibition has been examined and failed to improve RFS. However, the COMBI-AD study did demonstrate a substantial benefit for combination BRAF and MEK inhibition for patients with BRAF-mutated resected stage IIIA to IIIC melanoma.
Collapse
Affiliation(s)
- Ragini R Kudchadkar
- From the Department of Hematology/Oncology, Winship Cancer Institute, Emory University, Atlanta, GA; Department of Oncology, Lausanne University Hospital, Lausanne, Switzerland; Department of Surgical Oncology, Netherlands Cancer Institute, Antoni van Leeuwenhoek Hospital, Amsterdam, Netherlands
| | - Olivier Michielin
- From the Department of Hematology/Oncology, Winship Cancer Institute, Emory University, Atlanta, GA; Department of Oncology, Lausanne University Hospital, Lausanne, Switzerland; Department of Surgical Oncology, Netherlands Cancer Institute, Antoni van Leeuwenhoek Hospital, Amsterdam, Netherlands
| | - Alexander C J van Akkooi
- From the Department of Hematology/Oncology, Winship Cancer Institute, Emory University, Atlanta, GA; Department of Oncology, Lausanne University Hospital, Lausanne, Switzerland; Department of Surgical Oncology, Netherlands Cancer Institute, Antoni van Leeuwenhoek Hospital, Amsterdam, Netherlands
| |
Collapse
|
965
|
Amaria RN, Reddy SM, Tawbi HA, Davies MA, Ross MI, Glitza IC, Cormier JN, Lewis C, Hwu WJ, Hanna E, Diab A, Wong MK, Royal R, Gross N, Weber R, Lai SY, Ehlers R, Blando J, Milton DR, Woodman S, Kageyama R, Wells DK, Hwu P, Patel SP, Lucci A, Hessel A, Lee JE, Gershenwald J, Simpson L, Burton EM, Posada L, Haydu L, Wang L, Zhang S, Lazar AJ, Hudgens CW, Gopalakrishnan V, Reuben A, Andrews MC, Spencer CN, Prieto V, Sharma P, Allison J, Tetzlaff MT, Wargo JA. Neoadjuvant immune checkpoint blockade in high-risk resectable melanoma. Nat Med 2018; 24:1649-1654. [PMID: 30297909 PMCID: PMC6481682 DOI: 10.1038/s41591-018-0197-1] [Citation(s) in RCA: 609] [Impact Index Per Article: 87.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 08/21/2018] [Indexed: 12/17/2022]
Abstract
Preclinical studies suggest that treatment with neoadjuvant immune checkpoint blockade is associated with enhanced survival and antigen-specific T cell responses compared with adjuvant treatment1; however, optimal regimens have not been defined. Here we report results from a randomized phase 2 study of neoadjuvant nivolumab versus combined ipilimumab with nivolumab in 23 patients with high-risk resectable melanoma ( NCT02519322 ). RECIST overall response rates (ORR), pathologic complete response rates (pCR), treatment-related adverse events (trAEs) and immune correlates of response were assessed. Treatment with combined ipilimumab and nivolumab yielded high response rates (RECIST ORR 73%, pCR 45%) but substantial toxicity (73% grade 3 trAEs), whereas treatment with nivolumab monotherapy yielded modest responses (ORR 25%, pCR 25%) and low toxicity (8% grade 3 trAEs). Immune correlates of response were identified, demonstrating higher lymphoid infiltrates in responders to both therapies and a more clonal and diverse T cell infiltrate in responders to nivolumab monotherapy. These results describe the feasibility of neoadjuvant immune checkpoint blockade in melanoma and emphasize the need for additional studies to optimize treatment regimens and to validate putative biomarkers.
Collapse
Affiliation(s)
- Rodabe N. Amaria
- Department of Melanoma Medical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Sangeetha M. Reddy
- Department of Breast Medical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Hussein A. Tawbi
- Department of Melanoma Medical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Michael A. Davies
- Department of Melanoma Medical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Merrick I. Ross
- Department of Surgical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Isabella C. Glitza
- Department of Melanoma Medical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Janice N. Cormier
- Department of Surgical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Carol Lewis
- Department of Head and Neck Surgery, MD Anderson Cancer Center, Houston, Texas, USA
| | - Wen-Jen Hwu
- Department of Melanoma Medical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Ehab Hanna
- Department of Head and Neck Surgery, MD Anderson Cancer Center, Houston, Texas, USA
| | - Adi Diab
- Department of Melanoma Medical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Michael K. Wong
- Department of Melanoma Medical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Richard Royal
- Department of Surgical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Neil Gross
- Department of Head and Neck Surgery, MD Anderson Cancer Center, Houston, Texas, USA
| | - Randal Weber
- Department of Head and Neck Surgery, MD Anderson Cancer Center, Houston, Texas, USA
| | - Stephen Y. Lai
- Department of Head and Neck Surgery, MD Anderson Cancer Center, Houston, Texas, USA
| | - Richard Ehlers
- Department of Surgical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Jorge Blando
- Department of Immunology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Denái R. Milton
- Department of Biostatistics, MD Anderson Cancer Center, Houston, Texas, USA
| | - Scott Woodman
- Department of Melanoma Medical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Robin Kageyama
- Parker Institute for Cancer Immunotherapy, San Francisco, California, USA
| | - Danny K. Wells
- Parker Institute for Cancer Immunotherapy, San Francisco, California, USA
| | - Patrick Hwu
- Department of Melanoma Medical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Sapna P. Patel
- Department of Melanoma Medical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Anthony Lucci
- Department of Surgical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Amy Hessel
- Department of Head and Neck Surgery, MD Anderson Cancer Center, Houston, Texas, USA
| | - Jeffrey E. Lee
- Department of Surgical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Jeffrey Gershenwald
- Department of Surgical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Lauren Simpson
- Department of Melanoma Medical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Elizabeth M. Burton
- Department of Surgical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Liberty Posada
- Department of Melanoma Medical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Lauren Haydu
- Department of Surgical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Linghua Wang
- Department of Genomic Medicine, MD Anderson Cancer Center, Houston, Texas, USA
| | - Shaojun Zhang
- Department of Genomic Medicine, MD Anderson Cancer Center, Houston, Texas, USA
| | | | | | | | - Alexandre Reuben
- Department of Surgical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Miles C. Andrews
- Department of Surgical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | | | - Victor Prieto
- Department of Immunology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Padmanee Sharma
- Department of Immunology, MD Anderson Cancer Center, Houston, Texas, USA
- Department of Genitourinary Cancers, MD Anderson Cancer Center, Houston, Texas, USA
| | - James Allison
- Department of Immunology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Michael T. Tetzlaff
- Department of Pathology, MD Anderson Cancer Center, Houston, Texas, USA
- Department of Translational and Molecular Pathology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Jennifer A. Wargo
- Department of Surgical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
- Department of Genomic Medicine, MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
966
|
Ziogas DE, Kyrochristos ID, Roukos DH. Discovering novel valid biomarkers and drugs in patient-centric genomic trials: the new epoch of precision surgical oncology. Drug Discov Today 2018; 23:1848-1872. [PMID: 30077778 DOI: 10.1016/j.drudis.2018.07.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Revised: 07/10/2018] [Accepted: 07/26/2018] [Indexed: 12/16/2022]
Abstract
Despite standardization of multimodal treatment and approval of several targeted drugs for resectable, non-metastatic cancer (M0 patients), intrinsic and acquired resistance and relapse rates remain high, even in early-stage aggressive tumors. Genome analysis could overcome these unmet needs. Our comprehensive review underlines the controversy on stable or spatiotemporally evolving clones as well as promising yet inconclusive data on genome-based biomarkers and drug development. We propose clinicogenomic trials in M0 patients for the validation of intratumor heterogeneity (ITH), circulating genomic subclones (cGSs) and intra-patient genomic heterogeneity (IPGH) as biomarkers and simultaneous discovery of novel oncotargets. This evidence-based strategy highlights the coming of precision surgical oncology with a future perspective of understanding and disrupting deregulated transcriptional networks.
Collapse
Affiliation(s)
- Demosthenes E Ziogas
- Centre for Biosystems and Genome Network Medicine, Ioannina University, Ioannina, Greece; Department of Surgery, 'G. Hatzikosta' General Hospital, Ioannina, Greece
| | - Ioannis D Kyrochristos
- Centre for Biosystems and Genome Network Medicine, Ioannina University, Ioannina, Greece; Department of Surgery, Ioannina University Hospital, Ioannina, Greece
| | - Dimitrios H Roukos
- Centre for Biosystems and Genome Network Medicine, Ioannina University, Ioannina, Greece; Department of Surgery, Ioannina University Hospital, Ioannina, Greece; Department of Systems Biology, Biomedical Research Foundation of the Academy of Athens (BRFAA), Athens, Greece.
| |
Collapse
|
967
|
Swetter SM, Tsao H, Bichakjian CK, Curiel-Lewandrowski C, Elder DE, Gershenwald JE, Guild V, Grant-Kels JM, Halpern AC, Johnson TM, Sober AJ, Thompson JA, Wisco OJ, Wyatt S, Hu S, Lamina T. Guidelines of care for the management of primary cutaneous melanoma. J Am Acad Dermatol 2018; 80:208-250. [PMID: 30392755 DOI: 10.1016/j.jaad.2018.08.055] [Citation(s) in RCA: 394] [Impact Index Per Article: 56.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 08/28/2018] [Accepted: 08/29/2018] [Indexed: 12/12/2022]
Abstract
The incidence of primary cutaneous melanoma continues to increase each year. Melanoma accounts for the majority of skin cancer-related deaths, but treatment is usually curative following early detection of disease. In this American Academy of Dermatology clinical practice guideline, updated treatment recommendations are provided for patients with primary cutaneous melanoma (American Joint Committee on Cancer stages 0-IIC and pathologic stage III by virtue of a positive sentinel lymph node biopsy). Biopsy techniques for a lesion that is clinically suggestive of melanoma are reviewed, as are recommendations for the histopathologic interpretation of cutaneous melanoma. The use of laboratory, molecular, and imaging tests is examined in the initial work-up of patients with newly diagnosed melanoma and for follow-up of asymptomatic patients. With regard to treatment of primary cutaneous melanoma, recommendations for surgical margins and the concepts of staged excision (including Mohs micrographic surgery) and nonsurgical treatments for melanoma in situ, lentigo maligna type (including topical imiquimod and radiation therapy), are updated. The role of sentinel lymph node biopsy as a staging technique for cutaneous melanoma is described, with recommendations for its use in clinical practice. Finally, current data regarding pregnancy and melanoma, genetic testing for familial melanoma, and management of dermatologic toxicities related to novel targeted agents and immunotherapies for patients with advanced disease are summarized.
Collapse
Affiliation(s)
- Susan M Swetter
- Department of Dermatology, Stanford University Medical Center and Cancer Institute, Stanford, California; Veterans Affairs Palo Alto Health Care System, Palo Alto, California.
| | - Hensin Tsao
- Department of Dermatology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts; Wellman Center for Photomedicine, Boston, Massachusetts
| | - Christopher K Bichakjian
- Department of Dermatology, University of Michigan Health System, Ann Arbor, Michigan; Comprehensive Cancer Center, Ann Arbor, Michigan
| | - Clara Curiel-Lewandrowski
- Division of Dermatology, University of Arizona, Tucson, Arizona; University of Arizona Cancer Center, Tucson, Arizona
| | - David E Elder
- Department of Dermatology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania; Department of Pathology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jeffrey E Gershenwald
- Department of Surgical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas; Department of Cancer Biology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | | | - Jane M Grant-Kels
- Department of Dermatology, University of Connecticut Health Center, Farmington, Connecticut; Department of Pathology, University of Connecticut Health Center, Farmington, Connecticut; Department of Pediatrics, University of Connecticut Health Center, Farmington, Connecticut
| | - Allan C Halpern
- Department of Dermatology, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Timothy M Johnson
- Department of Dermatology, University of Michigan Health System, Ann Arbor, Michigan; Comprehensive Cancer Center, Ann Arbor, Michigan
| | - Arthur J Sober
- Department of Dermatology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - John A Thompson
- Division of Oncology, University of Washington, Seattle, Washington; Seattle Cancer Care Alliance, Seattle, Washington
| | - Oliver J Wisco
- Department of Dermatology, Oregon Health and Science University, Portland, Oregon
| | | | - Shasa Hu
- Department of Dermatology, University of Miami Health System, Miami, Florida
| | - Toyin Lamina
- American Academy of Dermatology, Rosemont, Illinois
| |
Collapse
|
968
|
Indini A, Brecht I, Del Vecchio M, Sultan I, Signoroni S, Ferrari A. Cutaneous melanoma in adolescents and young adults. Pediatr Blood Cancer 2018; 65:e27292. [PMID: 29968969 DOI: 10.1002/pbc.27292] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 05/21/2018] [Accepted: 05/29/2018] [Indexed: 12/24/2022]
Abstract
Cutaneous melanoma is rare in children, but has greater incidence in adolescents and young adults (AYAs). Diagnosis may be challenging due to its rarity in these age groups. Few studies have specifically addressed the topic of AYA melanoma. Though young-age melanoma may have particular biological characteristics, available data suggest that its clinical history is similar to that of adults. However, advances in treatment of adult melanoma have not been reflected in the treatment of AYAs. There is no standard treatment, and access to clinical trials is difficult for AYAs. Further efforts are needed to overcome these issues by improving cooperation with experts on adult melanoma.
Collapse
Affiliation(s)
- Alice Indini
- Melanoma Medical Oncology Unit, Department of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Ines Brecht
- Department of Pediatric Hematology and Oncology, University of Tubingen, Tubingen, Germany
| | - Michele Del Vecchio
- Melanoma Medical Oncology Unit, Department of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Iyad Sultan
- Department of Pediatric Oncology, King Hussein Cancer Center, Amman, Jordan
| | - Stefano Signoroni
- Unit of Hereditary Digestive Tract Tumours, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Andrea Ferrari
- Pediatric Oncology Unit, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| |
Collapse
|
969
|
McKean MA, Amaria RN. Multidisciplinary treatment strategies in high-risk resectable melanoma: Role of adjuvant and neoadjuvant therapy. Cancer Treat Rev 2018; 70:144-153. [DOI: 10.1016/j.ctrv.2018.08.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 08/27/2018] [Accepted: 08/28/2018] [Indexed: 12/17/2022]
|
970
|
Delman KA, Wong SL. Completion Node Dissection After Sentinel Node Biopsy in Melanoma. JAMA Surg 2018; 153:1045-1046. [DOI: 10.1001/jamasurg.2018.1516] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Affiliation(s)
| | - Sandra L. Wong
- Department of Surgery, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire
- Department of Surgery, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| |
Collapse
|
971
|
Gonzalez A. Sentinel Lymph Node Biopsy: Past and Present Implications for the Management of Cutaneous Melanoma with Nodal Metastasis. Am J Clin Dermatol 2018; 19:24-30. [PMID: 30374897 PMCID: PMC6244615 DOI: 10.1007/s40257-018-0379-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Although significant progress has been made in the understanding of melanoma pathophysiology and therapy, patients with metastatic melanoma still have a poor prognosis. The management of regional nodes remains a matter of debate. By replacing elective lymph node dissection, sentinel lymph node biopsy has revolutionized the treatment of malignant melanoma. In this paper, the history of the procedure is traced, and the indication for completion lymphadenectomy after positive sentinel node biopsy is discussed in light of the recent studies that addressed this issue. The role of adjuvant therapies in the management of patients with stage III melanoma is also discussed.
Collapse
Affiliation(s)
- Abel Gonzalez
- Mohs Surgery Unit, Institute of Oncology Angel H. Roffo, University of Buenos Aires, Av San Martin 5481, Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina.
- Unit of Cutaneous Oncology and Mohs Surgery, Institute Alexander Fleming, Conesa 1003, Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina.
| |
Collapse
|
972
|
Tarhini A, Lin Y, Lin H, Rahman Z, Vallabhaneni P, Mendiratta P, Pingpank JF, Holtzman MP, Yusko EC, Rytlewski JA, Rao UNM, Ferris RL, Kirkwood JM. Neoadjuvant ipilimumab (3 mg/kg or 10 mg/kg) and high dose IFN-α2b in locally/regionally advanced melanoma: safety, efficacy and impact on T-cell repertoire. J Immunother Cancer 2018; 6:112. [PMID: 30352626 PMCID: PMC6199801 DOI: 10.1186/s40425-018-0428-5] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 10/10/2018] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Neoadjuvant immunotherapy utilizing novel combinations has the potential to transform the standard of care for locally/regionally advanced melanoma. We hypothesized that neoadjuvant ipilimumab in combination with high dose IFNα2b (HDI) is safe and associated with durable pathologic complete responses (pCR). METHODS Patients with locally/regionally advanced melanoma were randomized to ipilimumab 3 or 10 mg/kg × 4 doses bracketing definitive surgery, then every 12 weeks × 4. HDI was given concurrently. We evaluated the safety and efficacy of the combination with ipilimumab 3 or 10 mg/kg. The impact on T-cell fraction and clonality were investigated in tumor and blood. RESULTS Thirty patients (age 37-76), 15 each at 3 and 10 mg/kg, 18 male and 12 female were treated. Considering immune related adverse events (irAEs) of interest, more grade 3/4 irAEs were seen with ipilimumab 10 mg/kg versus 3 mg/kg (p = 0.042). Among 28 evaluable patients, 11 relapsed, of whom 5 died. Median follow-up for 17 patients who have not relapsed was 32 months. The radiologic preoperative response rate was 36% (95% CI, 21-54); 4 patients at ipilimumab 3 mg/kg and 6 at 10 mg/kg and 2 (at 10 mg/kg) later relapsed. The pCR was 32% (95% CI, 18-51); 5 patients at ipilimumab 3 mg/kg and 4 at 10 mg/kg and one (at 3 mg/kg) had a late relapse. In patients with pCR, T-cell fraction was significantly higher when measured in primary melanoma tumors (p = 0.033). Higher tumor T-cell clonality in primary tumor and more so following neoadjuvant therapy was significantly associated with improved relapse free survival. CONCLUSIONS Neoadjuvant ipilimumab-HDI was relatively safe and exhibited promising tumor response rates with an associated measurable impact on T-cell fraction and clonality. Most pCRs were durable supporting the value of pCR as a primary endpoint in neoadjuvant immunotherapy trials. TRIAL REGISTRATION ClinicalTrials.gov, NCT01608594 . Registered 31 May 2012.
Collapse
Affiliation(s)
- Ahmad Tarhini
- UPMC Hillman Cancer Center, Pittsburgh, USA. .,Department of Hematology and Oncology, Cleveland Clinic Taussig Cancer Institute and Case Comprehensive Cancer Center, 9500 Euclid Ave CA6-157, Cleveland, OH, 44195, USA.
| | - Yan Lin
- UPMC Hillman Cancer Center, Pittsburgh, USA
| | - Huang Lin
- UPMC Hillman Cancer Center, Pittsburgh, USA
| | | | | | - Prateek Mendiratta
- Department of Hematology and Oncology, Cleveland Clinic Taussig Cancer Institute and Case Comprehensive Cancer Center, 9500 Euclid Ave CA6-157, Cleveland, OH, 44195, USA
| | | | | | | | | | | | | | | |
Collapse
|
973
|
Kakadia S, Yarlagadda N, Awad R, Kundranda M, Niu J, Naraev B, Mina L, Dragovich T, Gimbel M, Mahmoud F. Mechanisms of resistance to BRAF and MEK inhibitors and clinical update of US Food and Drug Administration-approved targeted therapy in advanced melanoma. Onco Targets Ther 2018; 11:7095-7107. [PMID: 30410366 PMCID: PMC6200076 DOI: 10.2147/ott.s182721] [Citation(s) in RCA: 190] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Approximately 50% of melanomas harbor an activating BRAF mutation. Combined BRAF and MEK inhibitors such as dabrafenib and trametinib, vemurafenib and cobimetinib, and encorafenib and binimetinib are US Food and Drug Administration (FDA)-approved to treat patients with BRAFV600-mutated advanced melanoma. Both genetic and epigenetic alterations play a major role in resistance to BRAF inhibitors by reactivation of the MAPK and/or the PI3K–Akt pathways. The role of BRAF inhibitors in modulating the immunomicroenvironment and perhaps enhancing the efficacy of checkpoint inhibitors is gaining interest. This article provides a comprehensive review of mechanisms of resistance to BRAF and MEK inhibitors in melanoma and summarizes landmark trials that led to the FDA approval of BRAF and MEK inhibitors in metastatic melanoma.
Collapse
Affiliation(s)
- Sunilkumar Kakadia
- Department of Internal Medicine, Division of Hematology and Oncology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Naveen Yarlagadda
- Department of Internal Medicine, Division of Hematology and Oncology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Ramez Awad
- Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Madappa Kundranda
- TW Lewis Melanoma Center of Excellence, Banner MD Anderson Cancer Center, Gilbert, AZ, USA,
| | - Jiaxin Niu
- TW Lewis Melanoma Center of Excellence, Banner MD Anderson Cancer Center, Gilbert, AZ, USA,
| | - Boris Naraev
- TW Lewis Melanoma Center of Excellence, Banner MD Anderson Cancer Center, Gilbert, AZ, USA,
| | - Lida Mina
- TW Lewis Melanoma Center of Excellence, Banner MD Anderson Cancer Center, Gilbert, AZ, USA,
| | - Tomislav Dragovich
- TW Lewis Melanoma Center of Excellence, Banner MD Anderson Cancer Center, Gilbert, AZ, USA,
| | - Mark Gimbel
- TW Lewis Melanoma Center of Excellence, Banner MD Anderson Cancer Center, Gilbert, AZ, USA,
| | - Fade Mahmoud
- TW Lewis Melanoma Center of Excellence, Banner MD Anderson Cancer Center, Gilbert, AZ, USA,
| |
Collapse
|
974
|
Verver D, van Klaveren D, Franke V, van Akkooi ACJ, Rutkowski P, Keilholz U, Eggermont AMM, Nijsten T, Grünhagen DJ, Verhoef C. Development and validation of a nomogram to predict recurrence and melanoma-specific mortality in patients with negative sentinel lymph nodes. Br J Surg 2018; 106:217-225. [PMID: 30307046 PMCID: PMC6585628 DOI: 10.1002/bjs.10995] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 06/04/2018] [Accepted: 08/08/2018] [Indexed: 12/21/2022]
Abstract
Background Patients with melanoma and negative sentinel nodes (SNs) have varying outcomes, dependent on several prognostic factors. Considering all these factors in a prediction model might aid in identifying patients who could benefit from a personalized treatment strategy. The objective was to construct and validate a nomogram for recurrence and melanoma‐specific mortality (MSM) in patients with melanoma and negative SNs. Methods A total of 3220 patients with negative SNs were identified from a cohort of 4124 patients from four EORTC Melanoma Group centres who underwent sentinel lymph node biopsy. Prognostic factors for recurrence and MSM were studied with Cox regression analysis. Significant factors were incorporated in the models. Performance was assessed by discrimination (c‐index) and calibration in cross‐validation across the four centres. A nomogram was developed for graphical presentation. Results There were 3180 eligible patients. The final prediction model for recurrence and the calibrated model for MSM included three independent prognostic factors: ulceration, anatomical location and Breslow thickness. The c‐index was 0·74 for recurrence and 0·76 for the calibrated MSM model. Cross‐validation across the four centres showed reasonable model performance. A nomogram was developed based on these models. One‐third of the patients had a 5‐year recurrence probability of 8·2 per cent or less, and one‐third had a recurrence probability of 23·0 per cent or more. Conclusion A nomogram for predicting recurrence and MSM in patients with melanoma and negative SNs was constructed and validated. It could provide personalized estimates useful for tailoring surveillance strategies (reduce or increase intensity), and selection of patients for adjuvant therapy or clinical trials. Could personalize care
Collapse
Affiliation(s)
- D Verver
- Department of Surgical Oncology, Erasmus MC Cancer Institute, University Medical Centre, Rotterdam, the Netherlands
| | - D van Klaveren
- Medical Statistics, Department of Biomedical Data Sciences, Leiden University Medical Centre, Leiden, the Netherlands
| | - V Franke
- Department of Surgical Oncology, Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, the Netherlands
| | - A C J van Akkooi
- Department of Surgical Oncology, Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, the Netherlands
| | - P Rutkowski
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie Institute Cancer Centre, Warsaw, Poland
| | - U Keilholz
- Charité Comprehensive Cancer Centre, University of Medicine Berlin, Berlin, Germany
| | - A M M Eggermont
- Department of Surgical Oncology, Gustave Roussy Cancer Campus, Villejuif, France
| | - T Nijsten
- Department of Dermatology, Erasmus MC University Medical Centre, Rotterdam, the Netherlands
| | - D J Grünhagen
- Department of Surgical Oncology, Erasmus MC Cancer Institute, University Medical Centre, Rotterdam, the Netherlands
| | - C Verhoef
- Department of Surgical Oncology, Erasmus MC Cancer Institute, University Medical Centre, Rotterdam, the Netherlands
| |
Collapse
|
975
|
Neoadjuvant versus adjuvant ipilimumab plus nivolumab in macroscopic stage III melanoma. Nat Med 2018; 24:1655-1661. [PMID: 30297911 DOI: 10.1038/s41591-018-0198-0] [Citation(s) in RCA: 625] [Impact Index Per Article: 89.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 08/06/2018] [Indexed: 12/25/2022]
Abstract
Adjuvant ipilimumab (anti-CTLA-4) and nivolumab (anti-PD-1) both improve relapse-free survival of stage III melanoma patients1,2. In stage IV disease, the combination of ipilimumab + nivolumab is superior to ipilimumab alone and also appears to be more effective than nivolumab monotherapy3. Preclinical work suggests that neoadjuvant application of checkpoint inhibitors may be superior to adjuvant therapy4. To address this question and to test feasibility, 20 patients with palpable stage III melanoma were 1:1 randomized to receive ipilimumab 3 mg kg-1 and nivolumab 1 mg kg-1, as either four courses after surgery (adjuvant arm) or two courses before surgery and two courses postsurgery (neoadjuvant arm). Neoadjuvant therapy was feasible, with all patients undergoing surgery at the preplanned time point. However in both arms, 9/10 patients experienced one or more grade 3/4 adverse events. Pathological responses were achieved in 7/9 (78%) patients treated in the neoadjuvant arm. None of these patients have relapsed so far (median follow-up, 25.6 months). We found that neoadjuvant ipilimumab + nivolumab expand more tumor-resident T cell clones than adjuvant application. While neoadjuvant therapy appears promising, with the current regimen it induced high toxicity rates; therefore, it needs further investigation to preserve efficacy but reduce toxicity.
Collapse
|
976
|
Brancaccio G, Napolitano S, Troiani T, Franco R, Iovino F, Reginelli A, Ciardiello F, Argenziano G. Eighth American Joint Committee on Cancer (AJCC) melanoma classification: what about stage IIC? Br J Dermatol 2018; 179:1422-1423. [DOI: 10.1111/bjd.17145] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- G. Brancaccio
- Dermatology Unit; Department of Internal & Experimental Medicine ‘F Magrassi’; University of Campania ‘Luigi Vanvitelli’; Via Pansini 5 80131 Naples Italy
| | - S. Napolitano
- Oncology Unit; Department of Internal & Experimental Medicine ‘F Magrassi’; University of Campania ‘Luigi Vanvitelli’; Via Pansini 5 80131 Naples Italy
| | - T. Troiani
- Oncology Unit; Department of Internal & Experimental Medicine ‘F Magrassi’; University of Campania ‘Luigi Vanvitelli’; Via Pansini 5 80131 Naples Italy
| | - R. Franco
- Pathology Unit; Department of Cardiothoracic and Respiratory Science; University of Campania ‘Luigi Vanvitelli’; Via Pansini 5 80131 Naples Italy
| | - F. Iovino
- Division of General Surgery; Department of Cardiothoracic and Respiratory Science; University of Campania ‘Luigi Vanvitelli’; Via Pansini 5 80131 Naples Italy
| | - A. Reginelli
- Department of Radiology & Radiotherapy, Department of Internal & Experimental Medicine ‘F Magrassi’; University of Campania ‘Luigi Vanvitelli’; Via Pansini 5 80131 Naples Italy
| | - F. Ciardiello
- Oncology Unit; Department of Internal & Experimental Medicine ‘F Magrassi’; University of Campania ‘Luigi Vanvitelli’; Via Pansini 5 80131 Naples Italy
| | - G. Argenziano
- Dermatology Unit; Department of Internal & Experimental Medicine ‘F Magrassi’; University of Campania ‘Luigi Vanvitelli’; Via Pansini 5 80131 Naples Italy
| |
Collapse
|
977
|
Hoang TT, Tanrikulu IC, Vatland QA, Hoang TM, Raines RT. A Human Ribonuclease Variant and ERK-Pathway Inhibitors Exhibit Highly Synergistic Toxicity for Cancer Cells. Mol Cancer Ther 2018; 17:2622-2632. [PMID: 30282811 DOI: 10.1158/1535-7163.mct-18-0724] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Revised: 08/15/2018] [Accepted: 09/27/2018] [Indexed: 12/20/2022]
Abstract
Pancreatic-type ribonucleases (ptRNases) are prevalent secretory enzymes that catalyze the cleavage of RNA. Ribonuclease inhibitor (RI) is a cytosolic protein that has femtomolar affinity for ptRNases, affording protection from the toxic catalytic activity of ptRNases, which can invade human cells. A human ptRNase variant that is resistant to inhibition by RI is a cytotoxin that is undergoing a clinical trial as a cancer chemotherapeutic agent. We find that the ptRNase and protein kinases in the ERK pathway exhibit strongly synergistic toxicity toward lung cancer cells (including a KRASG12C variant) and melanoma cells (including BRAFV600E variants). The synergism arises from inhibiting the phosphorylation of RI and thereby diminishing its affinity for the ptRNase. These findings link seemingly unrelated cellular processes, and suggest that the use of a kinase inhibitor to unleash a cytotoxic enzyme could lead to beneficial manifestations in the clinic.
Collapse
Affiliation(s)
- Trish T Hoang
- Department of Biochemistry, University of Wisconsin-Madison, Madison, Wisconsin
| | - I Caglar Tanrikulu
- Department of Biochemistry, University of Wisconsin-Madison, Madison, Wisconsin.,Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Quinn A Vatland
- Department of Biochemistry, University of Wisconsin-Madison, Madison, Wisconsin
| | - Trieu M Hoang
- Department of Biochemistry, University of Wisconsin-Madison, Madison, Wisconsin
| | - Ronald T Raines
- Department of Biochemistry, University of Wisconsin-Madison, Madison, Wisconsin. .,Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts.,Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin
| |
Collapse
|
978
|
Hieken TJ, Kane JM, Wong SL. The Role of Completion Lymph Node Dissection for Sentinel Lymph Node-Positive Melanoma. Ann Surg Oncol 2018; 26:1028-1034. [PMID: 30284132 DOI: 10.1245/s10434-018-6812-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2018] [Indexed: 12/12/2022]
Abstract
PURPOSE AND METHODS Completion lymph node dissection (CLND) for sentinel lymph node (SLN)-positive melanoma patients has been guideline-concordant standard of care since adoption of lymphatic mapping and SLN biopsy for the management of clinically node-negative melanoma patients more than 20 years ago. However, a trend for omission of CLND has been observed over the past decade, and we now have randomized, controlled clinical trial data to help guide treatment recommendations. Publication of these data prompted an American Society of Clinical Oncology-Society of Surgical Oncology 2018 clinical practice guideline update for these patients. RESULTS AND CONCLUSIONS Systematic review of current evidence supports a selective, individualized approach to CLND for SLN-positive melanoma. For low-risk, low-volume micrometastatic disease, SLN biopsy may be both diagnostic and therapeutic, and close clinical follow-up with imaging or CLND are reasonable options for appropriately selected patients. For higher-risk patients, omission of CLND requires careful consideration of risks versus benefits, relevant histopathology, and individualized patient discussion. This should address patient comorbidities and life expectancy, the predicted likelihood of additional positive nodes, availability of imaging surveillance, likelihood of adherence to imaging and clinical follow-up, consequences of regional recurrence, and the prognostic value of complete nodal staging and its impact on adjuvant therapy recommendations or clinical trial participation. Data on long-term outcomes, cost, and patient-reported quality of life measures are not yet available.
Collapse
Affiliation(s)
- Tina J Hieken
- Department of Surgery, Mayo Clinic, Rochester, MN, USA.
| | - John M Kane
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Sandra L Wong
- Department of Surgery, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| |
Collapse
|
979
|
Rowe SP, Luber B, Makell M, Brothers P, Santmyer J, Schollenberger MD, Quinn H, Edelstein DL, Jones FS, Bleich KB, Sharfman WH, Lipson EJ. From validity to clinical utility: the influence of circulating tumor DNA on melanoma patient management in a real-world setting. Mol Oncol 2018; 12:1661-1672. [PMID: 30113761 PMCID: PMC6165998 DOI: 10.1002/1878-0261.12373] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 06/25/2018] [Accepted: 08/05/2018] [Indexed: 01/08/2023] Open
Abstract
Melanoma currently lacks a reliable blood-based biomarker of disease activity, although circulating tumor DNA (ctDNA) may fill this role. We investigated the clinical utility (i.e., impact on clinical outcomes and interpretation of radiographic data) of measuring ctDNA in patients with metastatic or high-risk resected melanoma. Patients were prospectively accrued into ≥ 1 of three cohorts, as follows. Cohort A: patients with radiographically measurable metastatic melanoma who underwent comparison of ctDNA measured by a BEAMing digital PCR assay to tissue mutational status and total tumor burden; when appropriate, determinations about initiation of targeted therapy were based on ctDNA data. Cohorts B and C: patients with BRAF- or NRAS-mutant melanoma who had either undergone surgical resection of high-risk disease (cohort B) or were receiving or had received medical therapy for advanced disease (cohort C). Patients were followed longitudinally with serial ctDNA measurements with contemporaneous radiographic imaging to ascertain times to detection of disease activity and progressive disease, respectively. The sensitivity and specificity of the ctDNA assay were 86.8% and 100%, respectively. Higher tumor burden and visceral metastases were found to be associated with detectable ctDNA. In two patients in cohort A, ctDNA test results revealed a targetable mutation where tumor testing had not; both patients experienced a partial response to targeted therapy. In four of 30 patients with advanced melanoma, ctDNA assessments indicated evidence of melanoma activity that predicted radiographic evidence of disease progression by 8, 14, 25, and 38 weeks, respectively. CtDNA was detectable in three of these four patients coincident with radiographic evaluations that alone were interpreted as showing no evidence of neoplastic disease. Our findings provide evidence for the clinical utility of integrating ctDNA data in managing patients with melanoma in a real-world setting.
Collapse
Affiliation(s)
- Steven P. Rowe
- The Russell H. MorganDepartment of Radiology and Radiological ScienceJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Brandon Luber
- Division of Biostatistics and BioinformaticsDepartment of OncologySidney Kimmel Comprehensive Cancer CenterJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Monique Makell
- Department of OncologySidney Kimmel Comprehensive Cancer CenterJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Patricia Brothers
- Department of OncologySidney Kimmel Comprehensive Cancer CenterJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - JoAnn Santmyer
- Department of OncologySidney Kimmel Comprehensive Cancer CenterJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Megan D. Schollenberger
- Department of OncologySidney Kimmel Comprehensive Cancer CenterJohns Hopkins University School of MedicineBaltimoreMDUSA
| | | | | | | | - Karen B. Bleich
- Department of OncologySidney Kimmel Comprehensive Cancer CenterJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - William H. Sharfman
- Department of OncologySidney Kimmel Comprehensive Cancer CenterJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Evan J. Lipson
- Department of OncologySidney Kimmel Comprehensive Cancer CenterJohns Hopkins University School of MedicineBaltimoreMDUSA
| |
Collapse
|
980
|
Challenges and Opportunities of Neoadjuvant Treatment in Locally Advanced Melanoma. Am J Clin Dermatol 2018; 19:639-646. [PMID: 30039289 DOI: 10.1007/s40257-018-0371-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Locally advanced and metastatic melanoma have historically had poor survival outcomes. Long-term follow-up of both targeted therapies and immune checkpoint inhibitors has confirmed the survival benefit of these agents in stage IV melanoma, and recent studies have now demonstrated relapse-free survival benefits from these targeted and immunotherapeutic agents in the adjuvant setting. Neoadjuvant treatment of locally advanced melanoma, including in-transit disease, is now under investigation. Clinical trials have shown early promising results using either combination targeted therapy or immune checkpoint inhibitors. Neoadjuvant treatment may improve surgical morbidity, but balancing treatment efficacy and toxicity has already been challenging in the use of combination immune checkpoint inhibitors preoperatively. While improvement in relapse-free survival has been noted, additional follow-up of patients receiving neoadjuvant treatment will be necessary to report on long-term outcomes. Neoadjuvant treatment also provides additional translational research opportunities to determine predictive biomarkers for targeted therapy and immune checkpoint inhibitors. Evidence of early resistance to treatment may also lead to novel combination therapies to explore in future clinical trials. While neoadjuvant treatment in locally advanced melanoma has exciting potential, more investigation is necessary to determine efficacious regimens with manageable toxicities.
Collapse
|
981
|
Albertini MR, Yang RK, Ranheim EA, Hank JA, Zuleger CL, Weber S, Neuman H, Hartig G, Weigel T, Mahvi D, Henry MB, Quale R, McFarland T, Gan J, Carmichael L, Kim K, Loibner H, Gillies SD, Sondel PM. Pilot trial of the hu14.18-IL2 immunocytokine in patients with completely resectable recurrent stage III or stage IV melanoma. Cancer Immunol Immunother 2018; 67:1647-1658. [PMID: 30073390 PMCID: PMC6168354 DOI: 10.1007/s00262-018-2223-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 07/30/2018] [Indexed: 01/13/2023]
Abstract
Phase I testing of the hu14.18-IL2 immunocytokine (IC) in melanoma patients showed immune activation, reversible toxicities, and a maximal tolerated dose of 7.5 mg/m2/day. Preclinical data in IC-treated tumor-bearing mice with low tumor burden documented striking antitumor effects. Patients with completely resectable recurrent stage III or stage IV melanoma were scheduled to receive 3 courses of IC at 6 mg/m2/day i.v. on days 1, 2 and 3 of each 28-day course. Patients were randomized to complete surgical resection either following neoadjuvant (Group A) or prior to adjuvant (Group B) IC course 1. Primary objectives were to: (1) evaluate histological evidence of anti-tumor activity and (2) evaluate recurrence-free survival (RFS) and OS. Twenty melanoma patients were randomized to Group A (11 patients) or B (9 patients). Two Group B patients did not receive IC due to persistent disease following surgery. Six of 18 IC-treated patients remained free of recurrence, with a median RFS of 5.7 months (95% confidence interval (CI) 1.8-not reached). The 24-month RFS rate was 38.9% (95% CI 17.5-60.0%). The median follow-up of surviving patients was 50.0 months (range: 31.8-70.4). The 24-month OS rate was 65.0% (95% CI 40.3-81.5%). Toxicities were similar to those previously reported. Exploratory tumor-infiltrating lymphocyte (TIL) analyses suggest prognostic value of TILs from Group A patients. Prolonged tumor-free survival was seen in some melanoma patients at high risk for recurrence who were treated with IC.
Collapse
Affiliation(s)
- Mark R Albertini
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.
- Medical Service, William S. Middleton Memorial Veterans Hospital, Madison, WI, USA.
- University of Wisconsin Clinical Sciences Center, Room K6/530, 600 Highland Avenue, Madison, WI, 53792, USA.
| | - Richard K Yang
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Erik A Ranheim
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Jacquelyn A Hank
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Cindy L Zuleger
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Sharon Weber
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Heather Neuman
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Greg Hartig
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Tracey Weigel
- Westchester Medical Center, New York Medical College, Valhalla, New York, USA
| | - David Mahvi
- Department of Surgery, Medical University of South Carolina, Charleston, SC, USA
| | - Mary Beth Henry
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Renae Quale
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Thomas McFarland
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Jacek Gan
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Lakeesha Carmichael
- Department of Biostatistics and Medical Informatics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - KyungMann Kim
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Department of Biostatistics and Medical Informatics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | | | | | - Paul M Sondel
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| |
Collapse
|
982
|
Acral Melanoma: A Patient's Experience and Physician's Commentary. Dermatol Ther (Heidelb) 2018; 8:503-507. [PMID: 30229459 PMCID: PMC6261119 DOI: 10.1007/s13555-018-0260-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Indexed: 11/01/2022] Open
Abstract
This article, co-authored by a patient diagnosed with acral melanoma, discusses the patient's experience of being diagnosed with and treated with surgery for this disease. The physician discusses the epidemiology, genetics, diagnosis, treatment, and prognosis of acral melanoma. Follow-up care plans are also discussed.
Collapse
|
983
|
Masoud SJ, Perone JA, Farrow NE, Mosca PJ, Tyler DS, Beasley GM. Sentinel Lymph Node Biopsy and Completion Lymph Node Dissection for Melanoma. Curr Treat Options Oncol 2018; 19:55. [PMID: 30232648 PMCID: PMC6684152 DOI: 10.1007/s11864-018-0575-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OPINION STATEMENT This review critically evaluates recent trials which have challenged the practice of completion lymph node dissection (CLND) for melanoma patients diagnosed with regional metastasis by positive sentinel lymph node biopsy (SLNB). Two trials in the last 2 years, DeCOG-SLT and MSLT-II, found no significant differences in melanoma-specific survival between patients, whether they received immediate CLND or observation after positive SLNB, despite decreases in nodal recurrence achieved by dissection. These trials together disfavor routine CLND in most patients after positive SLNB. However, their conclusions are limited by study populations which overall harbored a lower burden of SLN disease. Special attention needs to be given to patients who do have higher risk disease, with SLN tumor burdens exceeding 1 mm in diameter, for whom CLND may remain both prognostic and therapeutic. Current guidelines thus recommend either CLND or careful observation after positive SLNB after appropriate risk stratification of patients. While a decline in CLND is inevitable, treatment of stage III melanoma is witnessing the concurrent rise of effective adjuvant therapies. PD-1 inhibitors such as nivolumab, or combination BRAF/MEK inhibitors for V600E or K mutant melanoma, which were previously available to only trial patients with completely resected stage III disease, are now approved for use in patients with positive SLNB alone. Providers are better equipped than ever to treat clinically occult, regional metastatic disease with SLNB followed by adjuvant therapy for most patients, but should take steps to avoid undertreatment of high-risk patients who may proceed to disease relapse or progression.
Collapse
Affiliation(s)
- Sabran J Masoud
- Department of Surgery, Duke University, Durham, NC, 27710, USA
| | - Jennifer A Perone
- Department of Surgery, University Texas Medical Branch, Galveston, TX, USA
| | - Norma E Farrow
- Department of Surgery, Duke University, Durham, NC, 27710, USA
| | - Paul J Mosca
- Department of Surgery, Duke University, Durham, NC, 27710, USA
| | - Douglas S Tyler
- Department of Surgery, University Texas Medical Branch, Galveston, TX, USA
| | - Georgia M Beasley
- Department of Surgery, Duke University, Durham, NC, 27710, USA.
- Duke University Medical Center, DUMC Box 3118, Durham, NC, 27710, USA.
| |
Collapse
|
984
|
Schadendorf D, van Akkooi ACJ, Berking C, Griewank KG, Gutzmer R, Hauschild A, Stang A, Roesch A, Ugurel S. Melanoma. Lancet 2018; 392:971-984. [PMID: 30238891 DOI: 10.1016/s0140-6736(18)31559-9] [Citation(s) in RCA: 957] [Impact Index Per Article: 136.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 06/07/2018] [Accepted: 06/29/2018] [Indexed: 02/08/2023]
Abstract
Cutaneous melanoma causes 55 500 deaths annually. The incidence and mortality rates of the disease differ widely across the globe depending on access to early detection and primary care. Once melanoma has spread, this type of cancer rapidly becomes life-threatening. For more than 40 years, few treatment options were available, and clinical trials during that time were all unsuccessful. Over the past 10 years, increased biological understanding and access to innovative therapeutic substances have transformed advanced melanoma into a new oncological model for treating solid cancers. Treatments that target B-Raf proto-oncogene serine/threonine-kinase (BRAF)V600 (Val600) mutations using selected BRAF inhibitors combined with mitogen-activated protein kinase inhibitors have significantly improved response and overall survival. Furthermore, advanced cutaneous melanoma has developed into a prototype for testing checkpoint-modulating agents, which has increased hope for long-term tumour containment and a potential cure. These expectations have been sustained by clinical success with targeted agents and antibodies that block programmed cell-death protein 1 in locoregional disease, which induces prolongation of relapse-free, distant-metastasis-free, and overall survival times.
Collapse
Affiliation(s)
- Dirk Schadendorf
- Department of Dermatology, University Hospital Essen, Essen, Germany; German Cancer Consortium, Heidelberg, Germany.
| | - Alexander C J van Akkooi
- Department of Surgical Oncology, Netherlands Cancer Institute Antoni van Leeuwenhoek, Amsterdam, Netherlands
| | - Carola Berking
- Department of Dermatology and Allergy, University Hospital Munich, Munich, Germany
| | - Klaus G Griewank
- Department of Dermatology, University Hospital Essen, Essen, Germany; German Cancer Consortium, Heidelberg, Germany
| | - Ralf Gutzmer
- Department of Dermatology, Hannover Medical School, Skin Cancer Centre Hannover, Hannover, Germany
| | - Axel Hauschild
- Department of Dermatology, University Hospital, Kiel, Germany
| | - Andreas Stang
- Centre of Clinical Epidemiology, Institute of Medical Informatics, Biometry, and Epidemiology, University Hospital Essen, Essen, Germany; Department of Epidemiology, School of Public Health, Boston University, Boston, MA, USA
| | - Alexander Roesch
- Department of Dermatology, University Hospital Essen, Essen, Germany; German Cancer Consortium, Heidelberg, Germany
| | - Selma Ugurel
- Department of Dermatology, University Hospital Essen, Essen, Germany; German Cancer Consortium, Heidelberg, Germany
| |
Collapse
|
985
|
Dummer R, Ascierto PA, Gogas HJ, Arance A, Mandala M, Liszkay G, Garbe C, Schadendorf D, Krajsova I, Gutzmer R, Chiarion Sileni V, Dutriaux C, de Groot JWB, Yamazaki N, Loquai C, Moutouh-de Parseval LA, Pickard MD, Sandor V, Robert C, Flaherty KT. Overall survival in patients with BRAF-mutant melanoma receiving encorafenib plus binimetinib versus vemurafenib or encorafenib (COLUMBUS): a multicentre, open-label, randomised, phase 3 trial. Lancet Oncol 2018; 19:1315-1327. [PMID: 30219628 DOI: 10.1016/s1470-2045(18)30497-2] [Citation(s) in RCA: 405] [Impact Index Per Article: 57.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 06/25/2018] [Accepted: 06/26/2018] [Indexed: 12/21/2022]
Abstract
BACKGROUND Encorafenib plus binimetinib and encorafenib alone improved progression-free survival compared with vemurafenib in patients with BRAFV600-mutant melanoma in the COLUMBUS trial. Here, we report the results of the secondary endpoint of overall survival. METHODS COLUMBUS was a two-part, randomised, open-label, phase 3 study done at 162 hospitals in 28 countries. Eligible patients were aged at least 18 years with histologically confirmed, locally advanced, unresectable, or metastatic cutaneous melanoma, or unknown primary melanoma, BRAFV600E or BRAFV600K mutation, an Eastern Cooperative Oncology Group (ECOG) performance status of 0 or 1, and were treatment naive or had progressed on or after first-line immunotherapy. In part 1 of the study, patients were randomly assigned (1:1:1) by use of interactive response technology to receive oral encorafenib 450 mg once daily plus oral binimetinib 45 mg twice daily (encorafenib plus binimetinib group), oral encorafenib 300 mg once daily (encorafenib group), or oral vemurafenib 960 mg twice daily (vemurafenib group). Randomisation was stratified by the American Joint Committee on Cancer stage, ECOG performance status, and BRAF mutation status. The primary outcome of the trial, progression-free survival with encorafenib plus binimetinib versus vemurafenib, was reported previously. Here we present the prespecified interim overall survival analysis. Efficacy analyses were by intent to treat. Safety was analysed in patients who received at least one dose of study drug. Part 2 of the study was initiated at the request of the US Food and Drug Administration to better understand the contribution of binimetinib to the combination therapy by comparing encorafenib 300 mg once daily plus binimetinib 45 mg twice daily with encorafenib 300 mg once daily alone. Results of part 2 will be published separately. This trial is ongoing and is registered with ClinicalTrials.gov, number NCT01909453, and EudraCT, number 2013-001176-38. FINDINGS Between Dec 30, 2013, and April 10, 2015, 577 of 1345 screened patients were randomly assigned to receive encorafenib plus binimetinib (n=192), encorafenib (n=194), or vemurafenib (n=191). Median follow-up for overall survival was 36·8 months (95% CI 35·9-37·5). Median overall survival was 33·6 months (95% CI 24·4-39·2) with encorafenib plus binimetinib and 16·9 months (14·0-24·5) with vemurafenib (hazard ratio 0·61 [95% CI 0·47-0·79]; two-sided p<0·0001). The most common grade 3 or 4 adverse events did not change substantially from the first report; those seen in more than 5% of patients treated with encorafenib plus binimetinib were increased γ-glutamyltransferase (18 [9%] of 192 patients), increased blood creatine phosphokinase (14 [7%]), and hypertension (12 [6%]); those seen with encorafenib alone were palmar-plantar erythrodysaesthesia syndrome (26 [14%] of 192 patients), myalgia (19 [10%]), and arthralgia (18 [9%]); and with vemurafenib the most common grade 3 or 4 adverse event was arthralgia (11 [6%] of 186 patients). One death in the combination treatment group was considered by the investigator to be possibly related to treatment. INTERPRETATION The combination of encorafenib plus binimetinib provided clinically meaningful efficacy with good tolerability as shown by improvements in both progression-free survival and overall survival compared with vemurafenib. These data suggest that the combination of encorafenib plus binimetinib is likely to become an important therapeutic option in patients with BRAFV600-mutant melanoma. FUNDING Array BioPharma, Novartis.
Collapse
Affiliation(s)
- Reinhard Dummer
- Department of Dermatology, University Hospital Zürich Skin Cancer Center, Zürich, Switzerland.
| | - Paolo A Ascierto
- Melanoma Unit, Cancer Immunotherapy and Innovative Therapies, Istituto Nazionale Tumori IRCCS Fondazione Pascale, Naples, Italy
| | - Helen J Gogas
- Department of Internal Medicine, National and Kapodistrian University of Athens, Laikon Hospital, Athens, Greece
| | - Ana Arance
- Department of Medical Oncology, Hospital Clinic of Barcelona, Barcelona, Spain
| | - Mario Mandala
- Department of Oncology and Haematology, Papa Giovanni XXIII Cancer Center Hospital, Bergamo, Italy
| | - Gabriella Liszkay
- Department of Dermatology, National Institute of Oncology, Budapest, Hungary
| | - Claus Garbe
- Department of Dermatology, University Hospital Tuebingen, Tuebingen, Germany
| | - Dirk Schadendorf
- Department of Dermatology, University Hospital Essen, Essen, Germany; German Cancer Consortium, Heidelberg, Germany
| | - Ivana Krajsova
- Department of Dermato-oncology, University Hospital Prague and Charles University First Medical Faculty, Prague, Czech Republic
| | - Ralf Gutzmer
- Department of Dermatology and Allergy, Skin Cancer Center Hannover, Hannover Medical School, Hannover, Germany
| | | | - Caroline Dutriaux
- Department of Oncologic Dermatology, Centre Hospitalier Universitaire de Bordeaux, Hôpital Saint-André, Bordeaux Cédex, France
| | | | - Naoya Yamazaki
- Department of Dermatologic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Carmen Loquai
- Department of Dermatology, University Medical Center Mainz, Mainz, Germany
| | | | | | | | - Caroline Robert
- Service of Dermatology, Department of Medicine and Paris-Sud University, Gustave Roussy, Villejuif Cedex, France
| | | |
Collapse
|
986
|
Rand JG, Faries MB. Omitting Completion Dissection in Melanoma? Help is Available for Surgeons Coping Without Routine Dissection, But More Work is Needed. Ann Surg Oncol 2018; 25:3416-3418. [PMID: 30209726 DOI: 10.1245/s10434-018-6744-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Indexed: 12/13/2022]
Affiliation(s)
- Jamie Green Rand
- Department of Surgery, Cedars Sinai Medical Center, Los Angeles, CA, USA
| | - Mark B Faries
- Department Surgical Oncology, The Angeles Clinic and Research Institute, Los Angeles, CA, USA.
| |
Collapse
|
987
|
The impact of effective systemic therapies on surgery for stage IV melanoma. Eur J Cancer 2018; 103:24-31. [PMID: 30196107 DOI: 10.1016/j.ejca.2018.08.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 08/02/2018] [Indexed: 01/11/2023]
Abstract
INTRODUCTION The outcomes of patients with metastatic melanoma have significantly improved with the introduction of effective systemic therapies (ESTs). The role of surgery in the context of ESTs for stage IV melanoma is evolving. We sought to characterise the changing patterns of surgery and oncological outcomes in patients with stage IV melanoma treated before and after the establishment of ESTs. METHODS Patients undergoing surgical resection of stage IV melanoma were identified from our institutional database from 2003 to 2015. Patients were grouped into two cohorts, those referred before EST (2003-2007) and after EST (2011-2015). Clinicopathological variables, patterns of surgery and oncological outcomes in the two groups were compared. RESULTS A total of 138 patients underwent surgery for stage IV melanoma (n = 69 in each cohort). We observed no significant difference in the ratio of operations/patients performed. However, the pattern of operations altered, with a significant decrease in in-transit excisions (0.9% vs. 19.4%, p < 0.001) and an increase in abdominal metastasectomies (21.1% vs. 4.2%, p < 0.001), in the after-EST cohort. Novel indications for surgical intervention were noted in the after-EST cohort, with a significant increase in potentially curative operations for residual oligometastatic disease (15.9% vs. 4.3%, p = 0.045). Survival after surgery was prolonged in the after-EST cohort (median 16 months vs. 6 months, p < 0.001), with the stage at initial metastasectomy (stage 4a, hazard ratio [HR] 0.45 (0.28-0.73), p = 0.001) and treatment with immune checkpoint inhibitors (HR 0.38 (0.25-0.60), p < 0.001) associated with prolonged survival. DISCUSSION Surgery remains important in the management of stage IV melanoma, with evolving indications and patterns of intervention after the introduction of ESTs. The combination of judicious surgery and EST may improve oncological outcomes.
Collapse
|
988
|
Tyrrell H, Payne M. Combatting mucosal melanoma: recent advances and future perspectives. Melanoma Manag 2018; 5:MMT11. [PMID: 30459941 PMCID: PMC6240847 DOI: 10.2217/mmt-2018-0003] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 08/03/2018] [Indexed: 12/20/2022] Open
Abstract
Mucosal melanomas are a rare subtype of melanoma and are associated with a particularly poor prognosis. Due to the rarity of the diagnosis, and the pace with which the management of cutaneous melanoma has evolved over recent years, there is little good evidence to guide management and evidence-based clinical guidelines are still in development in the UK. In this review we provide an overview of the management of mucosal melanoma, highlighting the critical differences between cutaneous and mucosal melanomas, before examining recent advances in the systemic treatment of this disease and likely future directions.
Collapse
Affiliation(s)
- Helen Tyrrell
- Oxford Cancer Centre, Oxford University Hospitals NHS Foundation Trust Oxford, Oxford, United Kingdom of Great Britain & Northern Ireland
| | - Miranda Payne
- Oxford Cancer Centre, Oxford University Hospitals NHS Foundation Trust Oxford, Oxford, United Kingdom of Great Britain & Northern Ireland
| |
Collapse
|
989
|
Davis EJ, Johnson DB, Sosman JA, Chandra S. Melanoma: What do all the mutations mean? Cancer 2018; 124:3490-3499. [PMID: 29663336 PMCID: PMC6191351 DOI: 10.1002/cncr.31345] [Citation(s) in RCA: 129] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 02/06/2018] [Accepted: 02/13/2018] [Indexed: 12/28/2022]
Abstract
Melanoma is one of the most highly mutated malignancies, largely as a function of its generation through ultraviolet light and other mutational processes. The wide array of mutations in both "driver" and "passenger" genes can present a confusing array of data for practitioners, particularly within the context of the recent revolutions in targeted and immune therapy. Although mutations in BRAF V600 clearly confer sensitivity to BRAF and mitogen-activated protein kinase kinase (MEK) inhibitors, the clinical implications of most other mutations are less often discussed and understood. In this review, we provide an overview of the high-frequency genomic alterations and their prognostic and therapeutic relevance in melanoma.
Collapse
Affiliation(s)
- Elizabeth J. Davis
- Department of Medicine, Vanderbilt University Medical Center and Vanderbilt Ingram Cancer Center
| | - Douglas B. Johnson
- Department of Medicine, Vanderbilt University Medical Center and Vanderbilt Ingram Cancer Center
| | - Jeffrey A. Sosman
- Department of Medicine, Feinberg School of Medicine and Robert H. Lurie Comprehensive Cancer Center
| | - Sunandana Chandra
- Department of Medicine, Feinberg School of Medicine and Robert H. Lurie Comprehensive Cancer Center
| |
Collapse
|
990
|
Xu M, Casio M, Range DE, Sosa JA, Counter CM. Copper Chelation as Targeted Therapy in a Mouse Model of Oncogenic BRAF-Driven Papillary Thyroid Cancer. Clin Cancer Res 2018; 24:4271-4281. [PMID: 30065097 PMCID: PMC6125179 DOI: 10.1158/1078-0432.ccr-17-3705] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 04/09/2018] [Accepted: 05/16/2018] [Indexed: 12/13/2022]
Abstract
Purpose: Sixty percent of papillary thyroid cancers (PTC) have an oncogenic (V600E) BRAF mutation. Inhibitors of BRAF and its substrates MEK1/2 are showing clinical promise in BRAFV600E PTC. PTC progression can be decades long, which is challenging in terms of toxicity and cost. We previously found that MEK1/2 require copper (Cu) for kinase activity and can be inhibited with the well-tolerated and economical Cu chelator tetrathiomolybdate (TM). We therefore tested TM for antineoplastic activity in BRAFV600E -positive PTC.Experimental Design: The efficacy of TM alone and in combination with current standard-of-care lenvatinib and sorafenib or BRAF and MEK1/2 inhibitors vemurafenib and trametinib was examined in BRAFV600E-positive human PTC cell lines and a genetically engineered mouse PTC model.Results: TM inhibited MEK1/2 kinase activity and transformed growth of PTC cells. TM was as or more potent than lenvatinib and sorafenib and enhanced the antineoplastic activity of sorafenib and vemurafenib. Activated ERK2, a substrate of MEK1/2, overcame this effect, consistent with TM deriving its antineoplastic activity by inhibiting MEK1/2. Oral TM reduced tumor burden and vemurafenib in a BrafV600E -positive mouse model of PTC. This effect was ascribed to a reduction of Cu in the tumors. TM reduced P-Erk1/2 in mouse PTC tumors, whereas genetic reduction of Cu in developing tumors trended towards a survival advantage. Finally, TM as a maintenance therapy after cessation of vemurafenib reduced tumor volume in the aforementioned PTC mouse model.Conclusions: TM inhibits BRAFV600E -driven PTC through inhibition of MEK1/2, supporting clinical evaluation of chronic TM therapy for this disease. Clin Cancer Res; 24(17); 4271-81. ©2018 AACR.
Collapse
Affiliation(s)
- MengMeng Xu
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina
- Medical-Scientist Training Program, Duke University Medical Center, Durham, North Carolina
| | - Michael Casio
- Department of Biomedical Engineering, Duke Pratt School of Engineering, Durham, North Carolina
| | - Danielle E Range
- Department of Pathology, Duke University Medical Center, Durham, North Carolina
| | - Julie A Sosa
- Departments of Surgery and Medicine, Duke Cancer Institute and Duke Clinical Research Institute, Duke University Medical Center, Durham, North Carolina
| | - Christopher M Counter
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina.
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina
| |
Collapse
|
991
|
Abstract
Introduction Breakthroughs in targeted therapy have significantly improved outcomes for many patients with advanced melanoma, including those with BRAFV600 mutant disease. Targeted therapy for BRAFV600-mutant metastatic melanoma includes combinations of BRAF inhibitors and MEK inhibitors, which improve response rates and prolong progression-free survival (PFS) and overall survival (OS) in these patients. However, while durable responses have been observed, many patients develop acquired resistance to these drugs. Areas covered Recent clinical trial updates and ongoing studies with targeted therapy for BRAF-V600 mutant melanoma are reviewed. Expert opinion Although BRAF targeted therapy remains an effective treatment for BRAF-mutant for melanoma, ongoing trials are exploring combinations with other targeted therapeutics and immunotherapeutics to determine whether tumor responses can be prolonged, and these drugs are increasingly utilized in the neoadjuvant and adjuvant settings.
Collapse
Affiliation(s)
- Zeynep Eroglu
- The Department of Cutaneous Oncology, Moffitt Cancer Center & Research Institute, 10920 McKinley Dr, Tampa, FL, USA
| | - Alpaslan Ozgun
- The Department of Cutaneous Oncology, Moffitt Cancer Center & Research Institute, 10920 McKinley Dr, Tampa, FL, USA
| |
Collapse
|
992
|
Liu X, Zhong D. [Research Progress of Targeted Therapy for BRAF Mutation
in Advanced Non-small Cell Lung Cancer]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2018; 21:635-640. [PMID: 30172272 PMCID: PMC6105358 DOI: 10.3779/j.issn.1009-3419.2018.08.10] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
靶向治疗是驱动基因阳性晚期非小细胞肺癌(non-small cell lung cancer, NSCLC)的重要治疗手段之一。鼠类肉瘤病毒癌基因同源物B1(v-raf murine sar-coma viral oncogene homolog B1, BRAF)基因是继表皮生长因子受体(epidermal growth factor receptor, EGFR)基因突变、间变性淋巴瘤激酶(anaplastic lymphoma kinase, ALK)基因融合和ROS1基因重排之后,NSCLC又一个重要的驱动基因。BRAF V600E突变占BRAF基因突变的一半以上,是晚期NSCLC的潜在治疗靶点,本文主要对BRAF基因突变类型及相关靶向研究进展进行综述。。
Collapse
Affiliation(s)
- Xia Liu
- Department of Medical Oncology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Diansheng Zhong
- Department of Medical Oncology, Tianjin Medical University General Hospital, Tianjin 300052, China
| |
Collapse
|
993
|
Sarkisian S, Davar D. MEK inhibitors for the treatment of NRAS mutant melanoma. DRUG DESIGN DEVELOPMENT AND THERAPY 2018; 12:2553-2565. [PMID: 30154648 PMCID: PMC6108333 DOI: 10.2147/dddt.s131721] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Melanoma is increasing rapidly in incidence and prevalence, especially in younger females and older males. Treatment options have expanded beyond high-dose interleukin 2 and adoptive T-cell therapy to include inhibitors of immune checkpoints programmed death 1 (PD-1) and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and small molecular inhibitors of pathways activated in melanoma, in particular the mitogen-activated protein kinase (MAPK) pathway. PD-1/CTLA-4 inhibitors and inhibitors of MAPK such as BRAF/MEK inhibitors have significantly improved survival in both the metastatic and, more recently, adjuvant settings. In this review, we discuss the preclinical data, clinical development, and potential use of novel MEK inhibitor binemetinib, particularly in the setting of NRAS mutant melanoma.
Collapse
Affiliation(s)
- Saro Sarkisian
- Division of General Internal Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Diwakar Davar
- Division of Hematology-Oncology, Hillman Cancer Center and University of Pittsburgh, Pittsburgh, PA, USA,
| |
Collapse
|
994
|
Purdie KJ, Proby CM, Rizvi H, Griffin H, Doorbar J, Sommerlad M, Feltkamp MC, der Meijden EV, Inman GJ, South AP, Leigh IM, Harwood CA. The Role of Human Papillomaviruses and Polyomaviruses in BRAF-Inhibitor Induced Cutaneous Squamous Cell Carcinoma and Benign Squamoproliferative Lesions. Front Microbiol 2018; 9:1806. [PMID: 30154763 PMCID: PMC6102365 DOI: 10.3389/fmicb.2018.01806] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 07/18/2018] [Indexed: 12/19/2022] Open
Abstract
Background: Human papillomavirus (HPV) has long been proposed as a cofactor in the pathogenesis of cutaneous squamous cell carcinoma (cSCC). More recently, the striking clinico-pathological features of cSCCs that complicate treatment of metastatic melanoma with inhibitors targeting BRAF mutations (BRAFi) has prompted speculation concerning a pathogenic role for oncogenic viruses. Here, we investigate HPV and human polyomaviruses (HPyV) and correlate with clinical, histologic, and genetic features in BRAFi-associated cSCC. Materials and Methods: Patients receiving BRAFi treatment were recruited at Barts Health NHS Trust. HPV DNA was detected in microdissected frozen samples using reverse line probe technology and degenerate and nested PCR. HPV immunohistochemistry was performed in a subset of samples. Quantitative PCR was performed to determine the presence and viral load of HPyVs with affinity for the skin (HPyV6, HPyV7, HPyV9, MCPyV, and TSPyV). These data were correlated with previous genetic mutational analysis of H, K and NRAS, NOTCH1/2, TP53, CDKN2A, CARD11, CREBBP, TGFBR1/2. Chromosomal aberrations were profiled using single nucleotide polymorphism (SNP) arrays. Results: Forty-five skin lesions from seven patients treated with single agent vemurafenib in 2012–2013 were analyzed: 12 cSCC, 19 viral warts (VW), 2 actinic keratosis (AK), 5 verrucous keratosis/other squamoproliferative (VK/SP) lesions, one melanocytic lesion and 6 normal skin samples. Significant histologic features of viral infection were seen in 10/12 (83%) cSCC. HPV DNA was detected in 18/19 (95%) VW/SP, 9/12 (75%) cSCC, 4/5 (80%) SP, and 3/6 (50%) normal skin samples and in 1/12 cases assessed by immunohistochemistry. HPyV was co-detected in 22/30 (73%) of samples, usually at low viral load, with MCPyV and HPyV7 the most common. SNP arrays confirmed low levels of chromosomal abnormality and there was no significant correlation between HPV or HPyV detection and individual gene mutations or overall mutational burden. Conclusion: Despite supportive clinicopathologic evidence, the role for HPV and HPyV infection in the pathogenesis of BRAFi-induced squamoproliferative lesions remains uncertain. Synergistic oncogenic mechanisms are plausible although speculative. Nonetheless, with the prospect of a significant increase in the adjuvant use of these drugs, further research is justified and may provide insight into the pathogenesis of other BRAFi-associated malignancies.
Collapse
Affiliation(s)
- Karin J Purdie
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Charlotte M Proby
- Division of Cancer Research, School of Medicine, University of Dundee, Dundee, United Kingdom
| | - Hasan Rizvi
- Department of Pathology, Barts Health NHS Trust, London, United Kingdom
| | - Heather Griffin
- Division of Virology, Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - John Doorbar
- Division of Virology, Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - Mary Sommerlad
- Department of Dermatology, Barts Health NHS Trust, London, United Kingdom
| | - Mariet C Feltkamp
- Department of Medical Microbiology, Leiden University Medical Center, Leiden, Netherlands
| | - Els Van der Meijden
- Department of Medical Microbiology, Leiden University Medical Center, Leiden, Netherlands
| | - Gareth J Inman
- Division of Cancer Research, School of Medicine, University of Dundee, Dundee, United Kingdom
| | - Andrew P South
- Department of Dermatology and Cutaneous Biology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Irene M Leigh
- Institute of Dentistry, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Catherine A Harwood
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom.,Department of Dermatology, Barts Health NHS Trust, London, United Kingdom
| |
Collapse
|
995
|
Leong SP, Wu M, Lu Y, Torre DM, von Bakonyi A, Ospina AM, Newsom JD, Luckett WS, Soon CW, Kim KB, Kashani-Sabet M. Intraoperative Imaging with a Portable Gamma Camera May Reduce the False-Negative Rate for Melanoma Sentinel Lymph Node Surgery. Ann Surg Oncol 2018; 25:3326-3333. [PMID: 30105436 PMCID: PMC6437127 DOI: 10.1245/s10434-018-6685-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Indexed: 01/09/2023]
Abstract
Background Preoperative imaging and intraoperative gamma probe (GP) localization is standard for identifying sentinel lymph nodes (SLNs) in melanoma patients. The aim of this prospective Institutional Review Board-approved study was to investigate whether an intraoperative portable gamma camera (PGC) improves SLN detection over the GP. Methods Lymphoscintigraphy and single photon emission computed tomography/computed tomography were performed after injection of 99mTc-Tilmanocept in melanoma patients (≥ 18 years, Breslow thickness ≥ 1.0 mm). A GP was used to localize the SLNs in each basin, which was explored by the GP to ensure that the operative field was < 10% counts of the hottest SLN. The PGC was then used after a negative GP screening. Any residual hotspots identified by the PGC were considered as additional SLNs and were removed following the 10% rule. Results Preoperative imaging of 100 patients identified 138 SLN basins, with 306 SLNs being identified by conventional surgery. The PGC localized 89 additional SLNs in 54 patients. Thus, the PGC identified an additional 23% of SLNs [95% confidence interval (CI) 18–27%]. Four of these 89 SLNs showed micrometastasis in four patients, in two of whom the only tumor-positive SLN was identified by the PGC, preventing two false-negative cases. Thus, the null hypothesis that the PGC did not detect additional positive SLNs was rejected (p = 0.000). The overall SLN positive rate was 9.9% (39/395, 95% CI 6–12), and the overall patient positive rate was increased using the PGC, from 25 to 27% (27/100). Conclusions Intraoperative PGC imaging yielded additional SLNs in a significant number of patients over GP alone. Identification of these additional SLNs resulted in upstaging of four patients with two patients being converted from a negative to a positive status, thus, preventing two false-negative cases.
Collapse
Affiliation(s)
| | - Max Wu
- California Pacific Medical Center, San Francisco, CA, USA
| | - Ying Lu
- Stanford University, Stanford, CA, USA
| | - Donald M Torre
- California Pacific Medical Center, San Francisco, CA, USA
| | | | | | - James D Newsom
- California Pacific Medical Center, San Francisco, CA, USA
| | | | | | - Kevin B Kim
- California Pacific Medical Center, San Francisco, CA, USA
| | | |
Collapse
|
996
|
Persa OD, Knuever J, Mauch C, Schlaak M. Complete lymph node dissection or observation in melanoma patients with multiple positive sentinel lymph nodes: A single-center retrospective analysis. J Dermatol 2018; 45:1191-1194. [DOI: 10.1111/1346-8138.14577] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 06/27/2018] [Indexed: 01/15/2023]
Affiliation(s)
- Oana-Diana Persa
- Department of Dermatology and Venereology; University of Cologne; Cologne Germany
| | - Jana Knuever
- Department of Dermatology and Venereology; University of Cologne; Cologne Germany
| | - Cornelia Mauch
- Department of Dermatology and Venereology; University of Cologne; Cologne Germany
| | - Max Schlaak
- Department of Dermatology and Venereology; University of Cologne; Cologne Germany
| |
Collapse
|
997
|
Friedman EB, Ferguson PM, Thompson JF. When is surgery for metastatic melanoma still the most appropriate treatment option? Expert Rev Anticancer Ther 2018; 18:943-945. [DOI: 10.1080/14737140.2018.1508346] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Affiliation(s)
- Erica B. Friedman
- Melanoma Institute Australia, The University of Sydney, North Sydney, Australia
| | - Peter M. Ferguson
- Melanoma Institute Australia, The University of Sydney, North Sydney, Australia
- Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Camperdown, Australia
- The Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - John F. Thompson
- Melanoma Institute Australia, The University of Sydney, North Sydney, Australia
- Department of Melanoma and Surgical Oncology, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
- The Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
998
|
Ben-Betzalel G, Baruch EN, Boursi B, Steinberg-Silman Y, Asher N, Shapira-Frommer R, Schachter J, Markel G. Possible immune adverse events as predictors of durable response to BRAF inhibitors in patients with BRAF V600-mutant metastatic melanoma. Eur J Cancer 2018; 101:229-235. [PMID: 30096703 DOI: 10.1016/j.ejca.2018.06.030] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Revised: 06/08/2018] [Accepted: 06/22/2018] [Indexed: 10/28/2022]
Abstract
BRAF inhibitors (BRAFi) and MEK inhibitors (MEKi) are among the cornerstones of metastatic melanoma therapy demonstrating excellent response rates with duration of 7-12 m. Long-term benefit from these agents was reported in patients with normal lactate dehydrogenase (LDH) and less than three disease sites. However, a treatment-dependent marker for long-term efficacy is lacking. Data suggest that immune-related adverse events (irAEs) are associated with clinical benefit in patients treated with immunotherapy and that response to BRAF/MEK therapy may have an underlying immune mechanism. We hypothesised that AEs with an underlying immune mechanism may be associated with a durable response to targeted therapy. We retrospectively identified a cohort of 78 BRAF V600-mutant metastatic melanoma patients treated with BRAFi or BRAFi + MEKi between November 2010 and November 2013. Four treatment-related AEs including vitiligo, uveitis, erythema nodosum and keratitis sicca were defined as irAEs of interest. Retrospective analysis of AEs in relationship to progression-free survival (PFS), disease burden and LDH levels was performed. Median PFS (mPFS) for all patients was 7.5 months with responses ongoing in eight patients as of April 2017. Ten patients were identified with the AEs defined previously. Cox regression analysis revealed a very strong association between those AEs and PFS; mPFS was 42.8 m in patients with at least one AE versus 6.1 m in those without an AE (hazard ratio [HR] 0.22, p = 0.002). This association was independent of LDH levels and disease burden (HR 0.24, p = 0.035). This analysis demonstrates a strong association between immune AEs and durable response to targeted therapy and may provide a treatment-related biomarker to estimate the outcome of therapy.
Collapse
Affiliation(s)
- Guy Ben-Betzalel
- Ella Lemelbaum Institute for Immuno-Oncology, Sheba Medical Center, Ramat-Gan, Israel.
| | - Erez N Baruch
- Ella Lemelbaum Institute for Immuno-Oncology, Sheba Medical Center, Ramat-Gan, Israel; Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel Aviv University, Israel
| | - Ben Boursi
- Division of Oncology, Sheba Medical Center, Ramat-Gan, Israel
| | - Yael Steinberg-Silman
- Ella Lemelbaum Institute for Immuno-Oncology, Sheba Medical Center, Ramat-Gan, Israel
| | - Nethanel Asher
- Ella Lemelbaum Institute for Immuno-Oncology, Sheba Medical Center, Ramat-Gan, Israel
| | | | - Jacob Schachter
- Ella Lemelbaum Institute for Immuno-Oncology, Sheba Medical Center, Ramat-Gan, Israel; Department of Oncology, Sackler Faculty of Medicine, Tel Aviv University, Israel
| | - Gal Markel
- Ella Lemelbaum Institute for Immuno-Oncology, Sheba Medical Center, Ramat-Gan, Israel; Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel Aviv University, Israel; Talpiot Medical Leadership Program, Sheba Medical Center, Ramat-Gan, Israel
| |
Collapse
|
999
|
Depenni R, De Rosa F, Greco S, Ridolfi L, Pellacani G, Ponti G, Cascinu S, Guidoboni M. Dabrafenib-trametinib combination in 'field-practice': an Italian experience. Future Oncol 2018; 14:2045-2052. [PMID: 30081673 DOI: 10.2217/fon-2017-0714] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
AIM This observational study investigates the effectiveness and safety of dabrafenib/trametinib combination in patients with metastatic melanoma. PATIENTS & METHODS Seventy-six patients treated with dabrafenib/trametinib (150 mg twice daily/2 mg once daily) were included. RESULTS Median progression-free survival was 9 months (95% CI: 7-11) and median overall survival was 14 months (11-16); disease control rate was 72%. Nine patients (12%) experienced a complete response. Of these, seven presented one metastatic site, none had lung or CNS metastasis, and none had elevated baseline lactate dehydrogenase (LDH) levels. Overall, subgroup analysis for patients with adverse prognostic features led to similar results. No new safety signals were reported. CONCLUSION Dabrafenib/trametinib combination can be effective and well-tolerated also in a heterogeneous 'real life' population comprising patients with adverse prognostic features.
Collapse
Affiliation(s)
- Roberta Depenni
- Division of Oncology, Department of Oncology & Hematology, University of Modena & Reggio Emilia, Modena, Italy
| | - Francesco De Rosa
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori, Meldola, Italy
| | - Stefano Greco
- Division of Oncology, Department of Oncology & Hematology, University of Modena & Reggio Emilia, Modena, Italy
| | - Laura Ridolfi
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori, Meldola, Italy
| | - Giovanni Pellacani
- Department of Dermatology, University of Modena & Reggio Emilia, Modena, Italy
| | - Giovanni Ponti
- Department of Diagnostic, Clinical Medicine & Public Health, Clinical Pathology Unit, University of Modena & Reggio Emilia, Modena, Italy
| | - Stefano Cascinu
- Division of Oncology, Department of Oncology & Hematology, University of Modena & Reggio Emilia, Modena, Italy
| | - Massimo Guidoboni
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori, Meldola, Italy
| |
Collapse
|
1000
|
Gastman BR, Gerami P, Kurley SJ, Cook RW, Leachman S, Vetto JT. Identification of patients at risk of metastasis using a prognostic 31-gene expression profile in subpopulations of melanoma patients with favorable outcomes by standard criteria. J Am Acad Dermatol 2018; 80:149-157.e4. [PMID: 30081113 DOI: 10.1016/j.jaad.2018.07.028] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 07/25/2018] [Accepted: 07/30/2018] [Indexed: 10/28/2022]
Abstract
BACKGROUND A substantial number of patients who relapse and die from cutaneous melanoma (CM) are categorized as being at low risk by traditional staging factors. The 31-gene expression profile (31-GEP) test independently stratifies metastatic risk of patients with CM as low (Class 1, with 1A indicating lowest risk) or high (Class 2,with 2B indicating highest risk). OBJECTIVE To assess risk prediction by the 31-GEP test within 3 low-risk (according to the American Joint Committee on Cancer) populations of patients with CM: those who are sentinel lymph node (SLN) negative, those with stage I to IIA tumors, and those with thin (≤1 mm [T1]) tumors. METHODS A total of 3 previous validation studies provided a nonoverlapping cohort of 690 patients with 31-GEP results, staging information, and survival outcomes. Kaplan-Meier and Cox regression analysis were performed. RESULTS The results included the identification of 70% of SLN-negative patients who experienced metastasis as Class 2, the discovery of reduced recurrence-free survival for patients with thin tumors and Class 2B biology compared with that of those with Class 1A biology (P < .0001); and determination of the 31-GEP test as an independent predictor of risk compared with traditional staging factors in patients with stage I to IIA tumors. LIMITATIONS Diagnoses spanned multiple versions of pathologic staging criteria. CONCLUSIONS The 31-GEP test identifies high-risk patients who are likely to experience recurrence or die of melanoma within low-risk groups of subpopulations of patients with CM who have SLN-negative disease, stage I to IIA tumors, and thin tumors.
Collapse
Affiliation(s)
- Brian R Gastman
- Department of Plastic Surgery, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio
| | - Pedram Gerami
- Department of Dermatology, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Skin Cancer Institute, Northwestern University Lurie Comprehensive Cancer Center, Chicago, Illinois
| | | | | | - Sancy Leachman
- Department of Dermatology, Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon
| | - John T Vetto
- Division of Surgical Oncology, Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon
| |
Collapse
|