851
|
Watanabe Y, Itoh M, Nakagawa H, Asahina A, Nobeyama Y. Role of interleukin‐24 in the tumor‐suppressive effects of interferon‐β on melanoma. Exp Dermatol 2019; 28:836-844. [DOI: 10.1111/exd.13955] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 04/27/2019] [Accepted: 05/02/2019] [Indexed: 12/19/2022]
Affiliation(s)
- Yoshinori Watanabe
- Department of Dermatology The Jikei University School of Medicine Tokyo Japan
| | - Munenari Itoh
- Department of Dermatology The Jikei University School of Medicine Tokyo Japan
| | - Hidemi Nakagawa
- Department of Dermatology The Jikei University School of Medicine Tokyo Japan
| | - Akihiko Asahina
- Department of Dermatology The Jikei University School of Medicine Tokyo Japan
| | - Yoshimasa Nobeyama
- Department of Dermatology The Jikei University School of Medicine Tokyo Japan
| |
Collapse
|
852
|
Pavri SN, Han G, Khan S, Han D. Does sentinel lymph node status have prognostic significance in patients with acral lentiginous melanoma? J Surg Oncol 2019; 119:1060-1069. [PMID: 30883783 DOI: 10.1002/jso.25445] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Revised: 02/20/2019] [Accepted: 02/26/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND The prognostic benefit of sentinel lymph node biopsy (SLNB) and factors predictive of survival specifically in patients with acral lentiginous melanoma (ALM) are unknown. METHODS The SEER database was queried for ALM cases that underwent SLNB from 1998 to 2013. Clinicopathological factors were correlated with SLN status, overall survival (OS), and melanoma-specific survival (MSS). RESULTS Median age for the 753 ALM study patients was 65 years, and 48.2% were male. Median thickness was 2 mm with 38.1% of cases having ulceration. SLN metastases were detected in 194 of 753 cases (25.7%). Multivariable analysis showed that thickness, Clark level IV-V, and ulceration significantly predicted for SLN metastasis (P < 0.05). For patients with positive SLN, 5-year OS and MSS were significantly worse at 48.1% and 58.9%, respectively, compared with 78.7% and 88.5%, respectively, for patients with negative SLN (P < 0.0001). On multivariable analyses, older age, male gender, increasing thickness, ulceration, and a positive SLN significantly predicted for worse OS and MSS (all P < 0.05). CONCLUSION This study confirms the important role of SLNB in ALM. SLN metastases are seen in 25.7% of ALM cases, providing significant prognostic information. In addition, thickness, ulceration status, and SLNB status significantly predict survival in patients with ALM.
Collapse
Affiliation(s)
- Sabrina N Pavri
- Aesthetic and Reconstructive Surgery Institute, UF Health Cancer Center-Orlando Health, Orlando, Florida
| | - Gang Han
- Department of Epidemiology and Biostatistics, School of Public Health, Texas A&M University, College Station, Texas
| | - Sajid Khan
- Section of Surgical Oncology, Department of Surgery, Yale University School of Medicine, New Haven, Connecticut
| | - Dale Han
- Division of Surgical Oncology, Department of Surgery, Oregon Health and Science University, Portland, Oregon
| |
Collapse
|
853
|
Lorentzen HF. Targeted therapy for malignant melanoma. Curr Opin Pharmacol 2019; 46:116-121. [PMID: 31261023 DOI: 10.1016/j.coph.2019.05.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 05/13/2019] [Accepted: 05/22/2019] [Indexed: 12/19/2022]
Abstract
Treatment of advanced melanoma has undergone a paradigm shift over the last 10-15 years. The frustrating results of studies on medical treatment ten years ago have been replaced by studies constantly improving survival in patients with advanced melanoma. Immune checkpoint inhibitors belong to one group of treatments and targeted therapy to another. Fifty percent of melanomas are BRAF mutation positive. Normally, the mitogen activated protein kinase or MAP kinase (Ras-BRAF-MEK-Erk chain) pathways translate external signals to intracellular growth and proliferation. In BRAF mutated melanoma cells, the mutated BRAF kinase is excessively active leading to autonomous proliferation and cancerous growth. This kinase can be blocked by BRAF-inhibitors. If given to BRAF negative melanoma patients, it may lead to disease progression because Ras is not inhibited in these cells. Development of Squamous cell carcinomas as a serious adverse event to BRAF inhibition may be caused by similar mechanisms in non BRAF mutated keratinocytes. A spontaneous and paradoxical loss of effect is seen with BRAF inhibitors due to various ways melanoma cells bypass BRAF. This is somewhat counteracted by the addition of a MEK1/2 inhibitor. Overall progression-free survival has increased from a median of two months for chemotherapy, via 7-8 months for BRAF inhibitor to 10-14 months for newer BRAF and MEK inhibitor combination therapy.
Collapse
Affiliation(s)
- Henrik F Lorentzen
- Department of Dermatology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 67, DK-8200 Aarhus N, Denmark.
| |
Collapse
|
854
|
Cook MG, Massi D, Szumera-Ciećkiewicz A, Van den Oord J, Blokx W, van Kempen LC, Balamurugan T, Bosisio F, Koljenović S, Portelli F, van Akkooi AC. An updated European Organisation for Research and Treatment of Cancer (EORTC) protocol for pathological evaluation of sentinel lymph nodes for melanoma. Eur J Cancer 2019; 114:1-7. [DOI: 10.1016/j.ejca.2019.03.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 02/13/2019] [Accepted: 03/10/2019] [Indexed: 10/27/2022]
|
855
|
Mason R, Au L, Ingles Garces A, Larkin J. Current and emerging systemic therapies for cutaneous metastatic melanoma. Expert Opin Pharmacother 2019; 20:1135-1152. [PMID: 31025594 DOI: 10.1080/14656566.2019.1601700] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 03/27/2019] [Indexed: 01/09/2023]
Abstract
INTRODUCTION Melanoma therapies have evolved rapidly, and initial successes have translated into survival gains for patients with advanced melanoma. Both targeted and immune-therapy now have evidence in earlier stage disease. There are many new agents and combinations of treatments in development as potential future treatment options. This highlights the need for a reflection on current treatment practice trends that are guiding the development of potential new therapies. AREAS COVERED In this review, the authors discuss the evidence for currently approved therapies for cutaneous melanoma, including adjuvant therapy, potential new biomarkers, and emerging treatments with early phase clinical trial data. The authors have searched both the PubMed and clinicaltrials.gov databases for published clinical trials and discuss selected landmark trials of current therapies and of investigational treatment strategies with early evidence for the treatment of melanoma. EXPERT OPINION Significant efficacy has been demonstrated with both immune checkpoint inhibitors and targeted therapies in treating advanced melanoma. A multitude of novel therapies are in development and there is need for instructive biomarker assessment to identify patients likely to respond or be refractory to current therapies, to identify mechanisms of resistance and to direct further treatment options to patients based on individual disease biology.
Collapse
Affiliation(s)
- Robert Mason
- a Clinical Research Fellow, Skin and Renal Units , The Royal Marsden Hospital , London , UK
- b Department of Medical Oncology , Gold Coast University Hospital , Southport , Queensland , Australia
| | - Lewis Au
- a Clinical Research Fellow, Skin and Renal Units , The Royal Marsden Hospital , London , UK
- c Division of Clinical Research , The Institute of Cancer Research , London , UK
| | - Alvaro Ingles Garces
- a Clinical Research Fellow, Skin and Renal Units , The Royal Marsden Hospital , London , UK
| | - James Larkin
- c Division of Clinical Research , The Institute of Cancer Research , London , UK
- d Consultant Oncologist , The Royal Marsden Hospital , London , UK
| |
Collapse
|
856
|
Hawkins ML, Rioth MJ, Eguchi MM, Cockburn M. Poor prognosis for thin ulcerated melanomas and implications for a more aggressive approach to treatment. J Am Acad Dermatol 2019; 80:1640-1649. [PMID: 30654077 PMCID: PMC7232854 DOI: 10.1016/j.jaad.2019.01.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 11/26/2018] [Accepted: 01/06/2019] [Indexed: 01/07/2023]
Abstract
BACKGROUND Clinical guidelines for the treatment of melanoma are based largely on the behavior of thicker tumors. As a result, little is known about survival differences among patients with thinner tumors. OBJECTIVE To investigate the variability in survival for American Joint Committee on Cancer stage T1 thin melanoma tumors, defined as tumors less than 1 mm thick at diagnosis. METHODS This population-based series included 43,008 non-Hispanic whites in whom cutaneous melanoma was diagnosed between 2004 and 2013 from the California Cancer Registry. Survival outcomes were estimated using the Kaplan-Meier method. Cox proportional hazard models were used to estimate risk of death. RESULTS Survival for patients with thin ulcerated tumors was comparable to that for patients with stage II tumors, who are currently treated more aggressively. At 12 months, patients with thin ulcerated tumors had approximately 6% lower survival (92.5% [95% confidence interval (CI), 90.6%-93.9%]) compared with patients with thin nonulcerated tumors (98.2% [95% CI, 98.0%-98.3%]). At 24 months, this survival difference increased (85.2% [95% CI, 82.8%-87.4%] vs 96.1% [95% CI, 95.8-96.3%] for those with thin ulcerated and thin nonulcerated tumors, respectively) and a greater than 15% survival difference was seen at 60 months. LIMITATIONS Previous reports of cancer registry data have noted some evidence of miscoding of thin tumors. CONCLUSION The poorer survival in patients with ulcerated tumors less than 1 mm thick implies the need for additional studies to determine potential benefits of more aggressive treatment.
Collapse
Affiliation(s)
- Makenzie L Hawkins
- University of Colorado Cancer Center, University of Colorado, Aurora, Colorado
| | - Matthew J Rioth
- University of Colorado Cancer Center, University of Colorado, Aurora, Colorado; Department of Medicine-Biomedical Informatics and Personalized Medicine, School of Medicine, University of Colorado, Aurora, Colorado
| | - Megan M Eguchi
- University of Colorado Cancer Center, University of Colorado, Aurora, Colorado
| | - Myles Cockburn
- University of Colorado Cancer Center, University of Colorado, Aurora, Colorado; Department of Dermatology, School of Medicine, University of Colorado, Aurora, Colorado; Department of Preventive Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California; Department of Dermatology, Keck School of Medicine of the University of Southern California, Los Angeles, California.
| |
Collapse
|
857
|
Meng D, Carvajal RD. KIT as an Oncogenic Driver in Melanoma: An Update on Clinical Development. Am J Clin Dermatol 2019; 20:315-323. [PMID: 30707374 DOI: 10.1007/s40257-018-0414-1] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Metastatic melanoma is a heterogenous disease that has served as a model for the development of both targeted therapy and immunotherapy. KIT-mutated melanoma represents a rare subset, most commonly arising from acral, mucosal, and chronically sun-damaged skin. Additionally, KIT alterations are enriched in the triple wild-type subtype of cutaneous melanoma. Activating alterations of KIT-a transmembrane receptor tyrosine kinase important for cell development, growth, and differentiation-have been shown to be critical to oncogenesis across many tumor subtypes. Following the successes of BRAF-targeted therapy in melanoma and KIT-targeted therapy in gastrointestinal stromal tumors, small-molecule tyrosine kinase inhibitors targeting KIT have been examined in KIT-mutated melanoma. KIT inhibitors that have been investigated in relevant clinical trials in advanced melanoma include imatinib, sunitinib, dasatinib, and nilotinib. In these studies, selected patients with KIT-mutated melanoma were shown to be responsive to therapy with KIT inhibition, especially patients with L576P and K642E mutations. This has led to the incorporation of KIT-targeted therapy in the National Comprehensive Cancer Network guidelines for systemic therapy for metastatic or unresectable melanoma. Current research and development efforts include novel KIT-targeted therapies and testing KIT inhibitors in combination with immunotherapy.
Collapse
|
858
|
Rozeman EA, Menzies AM, van Akkooi ACJ, Adhikari C, Bierman C, van de Wiel BA, Scolyer RA, Krijgsman O, Sikorska K, Eriksson H, Broeks A, van Thienen JV, Guminski AD, Acosta AT, Ter Meulen S, Koenen AM, Bosch LJW, Shannon K, Pronk LM, Gonzalez M, Ch'ng S, Grijpink-Ongering LG, Stretch J, Heijmink S, van Tinteren H, Haanen JBAG, Nieweg OE, Klop WMC, Zuur CL, Saw RPM, van Houdt WJ, Peeper DS, Spillane AJ, Hansson J, Schumacher TN, Long GV, Blank CU. Identification of the optimal combination dosing schedule of neoadjuvant ipilimumab plus nivolumab in macroscopic stage III melanoma (OpACIN-neo): a multicentre, phase 2, randomised, controlled trial. Lancet Oncol 2019; 20:948-960. [PMID: 31160251 DOI: 10.1016/s1470-2045(19)30151-2] [Citation(s) in RCA: 360] [Impact Index Per Article: 60.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 03/08/2019] [Accepted: 03/14/2019] [Indexed: 12/22/2022]
Abstract
BACKGROUND The outcome of patients with macroscopic stage III melanoma is poor. Neoadjuvant treatment with ipilimumab plus nivolumab at the standard dosing schedule induced pathological responses in a high proportion of patients in two small independent early-phase trials, and no patients with a pathological response have relapsed after a median follow up of 32 months. However, toxicity of the standard ipilimumab plus nivolumab dosing schedule was high, preventing its broader clinical use. The aim of the OpACIN-neo trial was to identify a dosing schedule of ipilimumab plus nivolumab that is less toxic but equally effective. METHODS OpACIN-neo is a multicentre, open-label, phase 2, randomised, controlled trial. Eligible patients were aged at least 18 years, had a WHO performance status of 0-1, had resectable stage III melanoma involving lymph nodes only, and measurable disease according to the Response Evaluation Criteria in Solid Tumors version 1.1. Patients were enrolled from three medical centres in Australia, Sweden, and the Netherlands, and were randomly assigned (1:1:1), stratified by site, to one of three neoadjuvant dosing schedules: group A, two cycles of ipilimumab 3 mg/kg plus nivolumab 1 mg/kg once every 3 weeks intravenously; group B, two cycles of ipilimumab 1 mg/kg plus nivolumab 3 mg/kg once every 3 weeks intravenously; or group C, two cycles of ipilimumab 3 mg/kg once every 3 weeks directly followed by two cycles of nivolumab 3 mg/kg once every 2 weeks intravenously. The investigators, site staff, and patients were aware of the treatment assignment during the study participation. Pathologists were masked to treatment allocation and all other data. The primary endpoints were the proportion of patients with grade 3-4 immune-related toxicity within the first 12 weeks and the proportion of patients achieving a radiological objective response and pathological response at 6 weeks. Analyses were done in all patients who received at least one dose of study drug. This trial is registered with ClinicalTrials.gov, number NCT02977052, and is ongoing with an additional extension cohort and to complete survival analysis. FINDINGS Between Nov 24, 2016 and June 28, 2018, 105 patients were screened for eligibility, of whom 89 (85%) eligible patients were enrolled and randomly assigned to one of the three groups. Three patients were excluded after randomisation because they were found to be ineligible, and 86 received at least one dose of study drug; 30 patients in group A, 30 in group B, and 26 in group C (accrual to this group was closed early upon advice of the Data Safety Monitoring Board on June 4, 2018 because of severe adverse events). Within the first 12 weeks, grade 3-4 immune-related adverse events were observed in 12 (40%) of 30 patients in group A, six (20%) of 30 in group B, and 13 (50%) of 26 in group C. The difference in grade 3-4 toxicity between group B and A was -20% (95% CI -46 to 6; p=0·158) and between group C and group A was 10% (-20 to 40; p=0·591). The most common grade 3-4 adverse events were elevated liver enzymes in group A (six [20%)]) and colitis in group C (five [19%]); in group B, none of the grade 3-4 adverse events were seen in more than one patient. One patient (in group A) died 9·5 months after the start of treatment due to the consequences of late-onset immune-related encephalitis, which was possibly treatment-related. 19 (63% [95% CI 44-80]) of 30 patients in group A, 17 (57% [37-75]) of 30 in group B, and nine (35% [17-56]) of 26 in group C achieved a radiological objective response, while pathological responses occurred in 24 (80% [61-92]) patients in group A, 23 (77% [58-90]) in group B, and 17 (65% [44-83]) in group C. INTERPRETATION OpACIN-neo identified a tolerable neoadjuvant dosing schedule (group B: two cycles of ipilimumab 1 mg/kg plus nivolumab 3 mg/kg) that induces a pathological response in a high proportion of patients and might be suitable for broader clinical use. When more mature data confirm these early observations, this schedule should be tested in randomised phase 3 studies versus adjuvant therapies, which are the current standard-of-care systemic therapy for patients with stage III melanoma. FUNDING Bristol-Myers Squibb.
Collapse
Affiliation(s)
| | - Alexander M Menzies
- Melanoma Institute of Australia, The University of Sydney, Sydney, NSW, Australia; Royal North Shore and Mater Hospitals, Sydney, NSW, Australia
| | | | - Chandra Adhikari
- Melanoma Institute of Australia, The University of Sydney, Sydney, NSW, Australia
| | | | | | - Richard A Scolyer
- Melanoma Institute of Australia, The University of Sydney, Sydney, NSW, Australia; Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | | | | | - Hanna Eriksson
- Karolinska University Hospital and Karolinska Institute, Stockholm, Sweden
| | | | | | - Alexander D Guminski
- Melanoma Institute of Australia, The University of Sydney, Sydney, NSW, Australia; Royal North Shore and Mater Hospitals, Sydney, NSW, Australia
| | | | | | | | | | - Kerwin Shannon
- Melanoma Institute of Australia, The University of Sydney, Sydney, NSW, Australia; Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Loes M Pronk
- The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Maria Gonzalez
- Melanoma Institute of Australia, The University of Sydney, Sydney, NSW, Australia
| | - Sydney Ch'ng
- Melanoma Institute of Australia, The University of Sydney, Sydney, NSW, Australia; Royal North Shore and Mater Hospitals, Sydney, NSW, Australia; Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | | | - Jonathan Stretch
- Melanoma Institute of Australia, The University of Sydney, Sydney, NSW, Australia; Royal North Shore and Mater Hospitals, Sydney, NSW, Australia; Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Stijn Heijmink
- The Netherlands Cancer Institute, Amsterdam, Netherlands
| | | | | | - Omgo E Nieweg
- Melanoma Institute of Australia, The University of Sydney, Sydney, NSW, Australia; Royal North Shore and Mater Hospitals, Sydney, NSW, Australia; Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | | | | | - Robyn P M Saw
- Melanoma Institute of Australia, The University of Sydney, Sydney, NSW, Australia; Royal North Shore and Mater Hospitals, Sydney, NSW, Australia; Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | | | | | - Andrew J Spillane
- Melanoma Institute of Australia, The University of Sydney, Sydney, NSW, Australia; Royal North Shore and Mater Hospitals, Sydney, NSW, Australia
| | - Johan Hansson
- Karolinska University Hospital and Karolinska Institute, Stockholm, Sweden
| | | | - Georgina V Long
- Melanoma Institute of Australia, The University of Sydney, Sydney, NSW, Australia
| | | |
Collapse
|
859
|
Dillman RO, Cornforth AN, McClay EF, Depriest C. Patient-specific dendritic cell vaccines with autologous tumor antigens in 72 patients with metastatic melanoma. Melanoma Manag 2019; 6:MMT20. [PMID: 31406564 PMCID: PMC6688559 DOI: 10.2217/mmt-2018-0010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Aim: Metastatic melanoma patients were treated with patient-specific vaccines consisting of autologous dendritic cells loaded with antigens from irradiated cells from short-term autologous tumor cell lines. Patients & methods: A total of 72 patients were enrolled in a single-arm Phase I/II (NCT00948480) trial or a randomized Phase II (NCT00436930). Results: Toxicity was minimal. Median overall survival (OS) was 49.4 months; 5-year OS 46%. A 5-year OS was 72% for 18 recurrent stage 3 without measurable disease when treated and 53% for 30 stage 4 without measurable disease when treated. A total of 24 patients with measurable stage 4 when treated (median of four prior therapies) had an 18.5 months median OS and 46% 2-year OS. Conclusion: This dendritic cell vaccine was associated with encouraging survival in all three clinical subsets. Clinicaltrial.gov NCT00436930 and NCT00948480.
Collapse
Affiliation(s)
- Robert O Dillman
- AIVITA Biomedical, Inc., Irvine, CA 92612, USA.,Hoag Cancer Institute, Newport Beach, CA 92658, USA.,University of California, Irvine, CA 92697, USA
| | | | - Edward F McClay
- California Cancer Associates for Research & Excellence (cCARE), Institute for Melanoma Research & Education, Encinitas, CA 92024, USA
| | | |
Collapse
|
860
|
Murali VS, Chang BJ, Fiolka R, Danuser G, Cobanoglu MC, Welf ES. An image-based assay to quantify changes in proliferation and viability upon drug treatment in 3D microenvironments. BMC Cancer 2019; 19:502. [PMID: 31138163 PMCID: PMC6537405 DOI: 10.1186/s12885-019-5694-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 05/08/2019] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Every biological experiment requires a choice of throughput balanced against physiological relevance. Most primary drug screens neglect critical parameters such as microenvironmental conditions, cell-cell heterogeneity, and specific readouts of cell fate for the sake of throughput. METHODS Here we describe a methodology to quantify proliferation and viability of single cells in 3D culture conditions by leveraging automated microscopy and image analysis to facilitate reliable and high-throughput measurements. We detail experimental conditions that can be adjusted to increase either throughput or robustness of the assay, and we provide a stand alone image analysis program for users who wish to implement this 3D drug screening assay in high throughput. RESULTS We demonstrate this approach by evaluating a combination of RAF and MEK inhibitors on melanoma cells, showing that cells cultured in 3D collagen-based matrices are more sensitive than cells grown in 2D culture, and that cell proliferation is much more sensitive than cell viability. We also find that cells grown in 3D cultured spheroids exhibit equivalent sensitivity to single cells grown in 3D collagen, suggesting that for the case of melanoma, a 3D single cell model may be equally effective for drug identification as 3D spheroids models. The single cell resolution of this approach enables stratification of heterogeneous populations of cells into differentially responsive subtypes upon drug treatment, which we demonstrate by determining the effect of RAK/MEK inhibition on melanoma cells co-cultured with fibroblasts. Furthermore, we show that spheroids grown from single cells exhibit dramatic heterogeneity to drug response, suggesting that heritable drug resistance can arise stochastically in single cells but be retained by subsequent generations. CONCLUSION In summary, image-based analysis renders cell fate detection robust, sensitive, and high-throughput, enabling cell fate evaluation of single cells in more complex microenvironmental conditions.
Collapse
Affiliation(s)
- Vasanth S. Murali
- Department of Cell Biology, UT Southwestern Medical Center, Dallas, TX USA
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX USA
| | - Bo-Jui Chang
- Department of Cell Biology, UT Southwestern Medical Center, Dallas, TX USA
| | - Reto Fiolka
- Department of Cell Biology, UT Southwestern Medical Center, Dallas, TX USA
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX USA
| | - Gaudenz Danuser
- Department of Cell Biology, UT Southwestern Medical Center, Dallas, TX USA
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX USA
| | - Murat Can Cobanoglu
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX USA
| | - Erik S. Welf
- Department of Cell Biology, UT Southwestern Medical Center, Dallas, TX USA
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX USA
| |
Collapse
|
861
|
[Reprint of: New guidelines for stage III melanoma (the French Cutaneous Oncology Group)]. Bull Cancer 2019; 106:560-573. [PMID: 31122657 DOI: 10.1016/j.bulcan.2019.04.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 01/15/2019] [Indexed: 11/22/2022]
Abstract
Improved knowledge of sentinel node procedures coupled with the results of adjuvant clinical trials in stage III melanoma have prompted the French Cutaneous Oncology Group to propose new guidelines for the management of stage III melanoma. These guidelines comply with the principles of the evidence-based medicine.
Collapse
|
862
|
Bernicker EH. What the Oncologist Needs From the Pathologist for Tyrosine Kinase Inhibitor Therapies. Arch Pathol Lab Med 2019; 143:1089-1092. [PMID: 31100016 DOI: 10.5858/arpa.2019-0210-sa] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Eric H Bernicker
- From Thoracic and Uveal Melanoma Medical Oncology, Cancer Center, Houston Methodist Hospital, Houston, Texas; and Clinical Medicine, Weill Cornell Medical College, New York, New York
| |
Collapse
|
863
|
Heinzerling L, Eigentler TK, Fluck M, Hassel JC, Heller-Schenck D, Leipe J, Pauschinger M, Vogel A, Zimmer L, Gutzmer R. Tolerability of BRAF/MEK inhibitor combinations: adverse event evaluation and management. ESMO Open 2019; 4:e000491. [PMID: 31231568 PMCID: PMC6555610 DOI: 10.1136/esmoopen-2019-000491] [Citation(s) in RCA: 155] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 03/06/2019] [Accepted: 03/31/2019] [Indexed: 12/11/2022] Open
Abstract
The inhibition of the mitogen-activated protein kinases signalling pathway through combined use of BRAF and MEK inhibitors (BRAFi+MEKi) represents an established therapeutic option in patients with BRAF-mutated, advanced melanoma. These efficient therapies are well tolerated with mostly moderate and reversible side effects and a discontinuation rate due to adverse events of 11.5%-15.7%. Median duration of therapy ranges between 8.8 and 11.7 months. Based on data from confirmatory trials, safety profiles of three BRAFi+MEKi combinations were reviewed, that is, dabrafenib plus trametinib, vemurafenib plus cobimetinib and encorafenib plus binimetinib. Many adverse events are class effects, such as cutaneous, gastrointestinal, ocular, cardiac and musculoskeletal events; some adverse events are substance associated. Fever (dabrafenib) and photosensitivity (vemurafenib) are the most common and clinically prominent examples. Other adverse events are less frequent and the association to one substance is less strong such as anaemia, facial paresis (encorafenib), neutropenia (dabrafenib), skin rash, QTc prolongation and increased liver function tests (vemurafenib). This narrative review provides recommendations for monitoring, adverse event evaluation and management focusing on the clinically relevant side effects of the three regimens.
Collapse
Affiliation(s)
- Lucie Heinzerling
- Department of Dermatology, University of Erlangen, Erlangen, Germany
| | - Thomas K Eigentler
- Department of Dermatology, Center for Dermatooncology, University Medical Center Tübingen, Tübingen, Germany
| | - Michael Fluck
- Department of Internal Medicine, Fachklinik Hornheide, Münster, Germany
| | - Jessica C Hassel
- Department of Dermatology, University Hospital Heidelberg, Heidelberg, Germany
| | | | - Jan Leipe
- Division of Rheumatology and Clinical Immunology, Medizinische Klinik und Poliklinik IV, University of Munich, Munich, Germany
| | - Matthias Pauschinger
- Department of Cardiology, Klinikum Nürnberg Süd, Paracelsus Medical University Nürnberg, Nuremberg, Germany
| | - Arndt Vogel
- Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Lisa Zimmer
- Department of Dermatology, University Hospital, University Essen-Duisburg, Essen, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Ralf Gutzmer
- Department of Dermatology, Skin Cancer Center Hannover, Hannover Medical School, Hannover, Germany
| |
Collapse
|
864
|
Huey RW, Hawk E, Offodile AC. Mind the Gap: Precision Oncology and Its Potential to Widen Disparities. J Oncol Pract 2019; 15:301-304. [PMID: 31112478 DOI: 10.1200/jop.19.00102] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Ryan W Huey
- 1 University of Texas MD Anderson Cancer Center, Houston, TX
| | - Ernest Hawk
- 1 University of Texas MD Anderson Cancer Center, Houston, TX
| | - Anaeze C Offodile
- 1 University of Texas MD Anderson Cancer Center, Houston, TX.,2 Baker Institute for Public Policy, Rice University, Houston, TX
| |
Collapse
|
865
|
Postsurgical treatment landscape and economic burden of locoregional and distant recurrence in patients with operable nonmetastatic melanoma. Melanoma Res 2019; 28:618-628. [PMID: 30216199 PMCID: PMC6221390 DOI: 10.1097/cmr.0000000000000507] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Supplemental Digital Content is available in the text. Surgery is the mainstay treatment for operable nonmetastatic melanoma, but recurrences are common and limit patients’ survival. This study aimed to describe real-world patterns of treatment and recurrence in patients with melanoma and to quantify healthcare resource utilization (HRU) and costs associated with episodes of locoregional/distant recurrences. Adults with nonmetastatic melanoma who underwent melanoma lymph node surgery were identified from the Truven Health MarketScan database (1 January 2008 to 31 July 2017). Locoregional and distant recurrence(s) were identified on the basis of postsurgery recurrence indicators (i.e. initiation of new melanoma pharmacotherapy, new radiotherapy, or new surgery; secondary malignancy diagnoses). Of 6400 eligible patients, 219 (3.4%) initiated adjuvant therapy within 3 months of surgery, mostly with interferon α-2b (n=206/219, 94.1%). A total of 1191/6400 (18.6%) patients developed recurrence(s) over a median follow-up of 23.1 months (102/6400, 1.6% distant recurrences). Among the 219 patients initiated on adjuvant therapy, 73 (33.3%) experienced recurrences (distant recurrences: 13/219, 5.9%). The mean total all-cause healthcare cost was $2645 per patient per month (PPPM) during locoregional recurrence episodes and $12 940 PPPM during distant recurrence episodes. In the year after recurrence, HRU was particularly higher in patients with distant recurrence versus recurrence-free matched controls: by 9.2 inpatient admissions, 54.4 inpatient days, 8.8 emergency department admissions, and 185.9 outpatient visits (per 100 person-months), whereas all-cause healthcare costs were higher by $14 953 PPPM. It remains to be determined whether the new generation of adjuvant therapies, such as immune checkpoint inhibitors and targeted agents, will increase the use of adjuvant therapies, and reduce the risk of recurrences and associated HRU/cost.
Collapse
|
866
|
Hindié E. What Is the Role of Dabrafenib Plus Trametinib Adjuvant Therapy in Stage IIIA Melanoma? J Clin Oncol 2019; 37:1355-1356. [DOI: 10.1200/jco.18.02075] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Elif Hindié
- Elif Hindié, MD, PhD, Bordeaux University Hospital, Bordeaux, France
| |
Collapse
|
867
|
Long GV, Dummer R, Schadendorf D, Santinami M, Atkinson V, Mandalà M, Chiarion-Sileni V, Larkin J, Nyakas M, Dutriaux C, Haydon A, Robert C, Mortier L, Schachter J, Lesimple T, Plummer R, Dasgupta K, Haas T, Shilkrut M, Gasal E, Kefford R, Kirkwood JM, Hauschild A. Reply to E. Hindié and K.R. Hess. J Clin Oncol 2019; 37:1356-1358. [DOI: 10.1200/jco.19.00004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Georgina V. Long
- Georgina V. Long, MD, PhD, Melanoma Institute Australia, The University of Sydney, and Royal North Shore and Mater Hospitals, Sydney, NSW, Australia; Reinhard Dummer, MD, University Hospital Zürich Skin Cancer Center, Zürich, Switzerland; Dirk Schadendorf, MD, PhD, University Hospital Essen, Essen, Germany, and German Cancer Consortium, Heidelberg, Germany; Mario Santinami, MD, Fondazione Istituto Nazionale Tumori, Milan, Italy; Victoria Atkinson, MD, Princess Alexandra Hospital, Gallipoli Medical
| | - Reinhard Dummer
- Georgina V. Long, MD, PhD, Melanoma Institute Australia, The University of Sydney, and Royal North Shore and Mater Hospitals, Sydney, NSW, Australia; Reinhard Dummer, MD, University Hospital Zürich Skin Cancer Center, Zürich, Switzerland; Dirk Schadendorf, MD, PhD, University Hospital Essen, Essen, Germany, and German Cancer Consortium, Heidelberg, Germany; Mario Santinami, MD, Fondazione Istituto Nazionale Tumori, Milan, Italy; Victoria Atkinson, MD, Princess Alexandra Hospital, Gallipoli Medical
| | - Dirk Schadendorf
- Georgina V. Long, MD, PhD, Melanoma Institute Australia, The University of Sydney, and Royal North Shore and Mater Hospitals, Sydney, NSW, Australia; Reinhard Dummer, MD, University Hospital Zürich Skin Cancer Center, Zürich, Switzerland; Dirk Schadendorf, MD, PhD, University Hospital Essen, Essen, Germany, and German Cancer Consortium, Heidelberg, Germany; Mario Santinami, MD, Fondazione Istituto Nazionale Tumori, Milan, Italy; Victoria Atkinson, MD, Princess Alexandra Hospital, Gallipoli Medical
| | - Mario Santinami
- Georgina V. Long, MD, PhD, Melanoma Institute Australia, The University of Sydney, and Royal North Shore and Mater Hospitals, Sydney, NSW, Australia; Reinhard Dummer, MD, University Hospital Zürich Skin Cancer Center, Zürich, Switzerland; Dirk Schadendorf, MD, PhD, University Hospital Essen, Essen, Germany, and German Cancer Consortium, Heidelberg, Germany; Mario Santinami, MD, Fondazione Istituto Nazionale Tumori, Milan, Italy; Victoria Atkinson, MD, Princess Alexandra Hospital, Gallipoli Medical
| | - Victoria Atkinson
- Georgina V. Long, MD, PhD, Melanoma Institute Australia, The University of Sydney, and Royal North Shore and Mater Hospitals, Sydney, NSW, Australia; Reinhard Dummer, MD, University Hospital Zürich Skin Cancer Center, Zürich, Switzerland; Dirk Schadendorf, MD, PhD, University Hospital Essen, Essen, Germany, and German Cancer Consortium, Heidelberg, Germany; Mario Santinami, MD, Fondazione Istituto Nazionale Tumori, Milan, Italy; Victoria Atkinson, MD, Princess Alexandra Hospital, Gallipoli Medical
| | - Mario Mandalà
- Georgina V. Long, MD, PhD, Melanoma Institute Australia, The University of Sydney, and Royal North Shore and Mater Hospitals, Sydney, NSW, Australia; Reinhard Dummer, MD, University Hospital Zürich Skin Cancer Center, Zürich, Switzerland; Dirk Schadendorf, MD, PhD, University Hospital Essen, Essen, Germany, and German Cancer Consortium, Heidelberg, Germany; Mario Santinami, MD, Fondazione Istituto Nazionale Tumori, Milan, Italy; Victoria Atkinson, MD, Princess Alexandra Hospital, Gallipoli Medical
| | - Vanna Chiarion-Sileni
- Georgina V. Long, MD, PhD, Melanoma Institute Australia, The University of Sydney, and Royal North Shore and Mater Hospitals, Sydney, NSW, Australia; Reinhard Dummer, MD, University Hospital Zürich Skin Cancer Center, Zürich, Switzerland; Dirk Schadendorf, MD, PhD, University Hospital Essen, Essen, Germany, and German Cancer Consortium, Heidelberg, Germany; Mario Santinami, MD, Fondazione Istituto Nazionale Tumori, Milan, Italy; Victoria Atkinson, MD, Princess Alexandra Hospital, Gallipoli Medical
| | - James Larkin
- Georgina V. Long, MD, PhD, Melanoma Institute Australia, The University of Sydney, and Royal North Shore and Mater Hospitals, Sydney, NSW, Australia; Reinhard Dummer, MD, University Hospital Zürich Skin Cancer Center, Zürich, Switzerland; Dirk Schadendorf, MD, PhD, University Hospital Essen, Essen, Germany, and German Cancer Consortium, Heidelberg, Germany; Mario Santinami, MD, Fondazione Istituto Nazionale Tumori, Milan, Italy; Victoria Atkinson, MD, Princess Alexandra Hospital, Gallipoli Medical
| | - Marta Nyakas
- Georgina V. Long, MD, PhD, Melanoma Institute Australia, The University of Sydney, and Royal North Shore and Mater Hospitals, Sydney, NSW, Australia; Reinhard Dummer, MD, University Hospital Zürich Skin Cancer Center, Zürich, Switzerland; Dirk Schadendorf, MD, PhD, University Hospital Essen, Essen, Germany, and German Cancer Consortium, Heidelberg, Germany; Mario Santinami, MD, Fondazione Istituto Nazionale Tumori, Milan, Italy; Victoria Atkinson, MD, Princess Alexandra Hospital, Gallipoli Medical
| | - Caroline Dutriaux
- Georgina V. Long, MD, PhD, Melanoma Institute Australia, The University of Sydney, and Royal North Shore and Mater Hospitals, Sydney, NSW, Australia; Reinhard Dummer, MD, University Hospital Zürich Skin Cancer Center, Zürich, Switzerland; Dirk Schadendorf, MD, PhD, University Hospital Essen, Essen, Germany, and German Cancer Consortium, Heidelberg, Germany; Mario Santinami, MD, Fondazione Istituto Nazionale Tumori, Milan, Italy; Victoria Atkinson, MD, Princess Alexandra Hospital, Gallipoli Medical
| | - Andrew Haydon
- Georgina V. Long, MD, PhD, Melanoma Institute Australia, The University of Sydney, and Royal North Shore and Mater Hospitals, Sydney, NSW, Australia; Reinhard Dummer, MD, University Hospital Zürich Skin Cancer Center, Zürich, Switzerland; Dirk Schadendorf, MD, PhD, University Hospital Essen, Essen, Germany, and German Cancer Consortium, Heidelberg, Germany; Mario Santinami, MD, Fondazione Istituto Nazionale Tumori, Milan, Italy; Victoria Atkinson, MD, Princess Alexandra Hospital, Gallipoli Medical
| | - Caroline Robert
- Georgina V. Long, MD, PhD, Melanoma Institute Australia, The University of Sydney, and Royal North Shore and Mater Hospitals, Sydney, NSW, Australia; Reinhard Dummer, MD, University Hospital Zürich Skin Cancer Center, Zürich, Switzerland; Dirk Schadendorf, MD, PhD, University Hospital Essen, Essen, Germany, and German Cancer Consortium, Heidelberg, Germany; Mario Santinami, MD, Fondazione Istituto Nazionale Tumori, Milan, Italy; Victoria Atkinson, MD, Princess Alexandra Hospital, Gallipoli Medical
| | - Laurent Mortier
- Georgina V. Long, MD, PhD, Melanoma Institute Australia, The University of Sydney, and Royal North Shore and Mater Hospitals, Sydney, NSW, Australia; Reinhard Dummer, MD, University Hospital Zürich Skin Cancer Center, Zürich, Switzerland; Dirk Schadendorf, MD, PhD, University Hospital Essen, Essen, Germany, and German Cancer Consortium, Heidelberg, Germany; Mario Santinami, MD, Fondazione Istituto Nazionale Tumori, Milan, Italy; Victoria Atkinson, MD, Princess Alexandra Hospital, Gallipoli Medical
| | - Jacob Schachter
- Georgina V. Long, MD, PhD, Melanoma Institute Australia, The University of Sydney, and Royal North Shore and Mater Hospitals, Sydney, NSW, Australia; Reinhard Dummer, MD, University Hospital Zürich Skin Cancer Center, Zürich, Switzerland; Dirk Schadendorf, MD, PhD, University Hospital Essen, Essen, Germany, and German Cancer Consortium, Heidelberg, Germany; Mario Santinami, MD, Fondazione Istituto Nazionale Tumori, Milan, Italy; Victoria Atkinson, MD, Princess Alexandra Hospital, Gallipoli Medical
| | - Thierry Lesimple
- Georgina V. Long, MD, PhD, Melanoma Institute Australia, The University of Sydney, and Royal North Shore and Mater Hospitals, Sydney, NSW, Australia; Reinhard Dummer, MD, University Hospital Zürich Skin Cancer Center, Zürich, Switzerland; Dirk Schadendorf, MD, PhD, University Hospital Essen, Essen, Germany, and German Cancer Consortium, Heidelberg, Germany; Mario Santinami, MD, Fondazione Istituto Nazionale Tumori, Milan, Italy; Victoria Atkinson, MD, Princess Alexandra Hospital, Gallipoli Medical
| | - Ruth Plummer
- Georgina V. Long, MD, PhD, Melanoma Institute Australia, The University of Sydney, and Royal North Shore and Mater Hospitals, Sydney, NSW, Australia; Reinhard Dummer, MD, University Hospital Zürich Skin Cancer Center, Zürich, Switzerland; Dirk Schadendorf, MD, PhD, University Hospital Essen, Essen, Germany, and German Cancer Consortium, Heidelberg, Germany; Mario Santinami, MD, Fondazione Istituto Nazionale Tumori, Milan, Italy; Victoria Atkinson, MD, Princess Alexandra Hospital, Gallipoli Medical
| | - Kohinoor Dasgupta
- Georgina V. Long, MD, PhD, Melanoma Institute Australia, The University of Sydney, and Royal North Shore and Mater Hospitals, Sydney, NSW, Australia; Reinhard Dummer, MD, University Hospital Zürich Skin Cancer Center, Zürich, Switzerland; Dirk Schadendorf, MD, PhD, University Hospital Essen, Essen, Germany, and German Cancer Consortium, Heidelberg, Germany; Mario Santinami, MD, Fondazione Istituto Nazionale Tumori, Milan, Italy; Victoria Atkinson, MD, Princess Alexandra Hospital, Gallipoli Medical
| | - Tomas Haas
- Georgina V. Long, MD, PhD, Melanoma Institute Australia, The University of Sydney, and Royal North Shore and Mater Hospitals, Sydney, NSW, Australia; Reinhard Dummer, MD, University Hospital Zürich Skin Cancer Center, Zürich, Switzerland; Dirk Schadendorf, MD, PhD, University Hospital Essen, Essen, Germany, and German Cancer Consortium, Heidelberg, Germany; Mario Santinami, MD, Fondazione Istituto Nazionale Tumori, Milan, Italy; Victoria Atkinson, MD, Princess Alexandra Hospital, Gallipoli Medical
| | - Mark Shilkrut
- Georgina V. Long, MD, PhD, Melanoma Institute Australia, The University of Sydney, and Royal North Shore and Mater Hospitals, Sydney, NSW, Australia; Reinhard Dummer, MD, University Hospital Zürich Skin Cancer Center, Zürich, Switzerland; Dirk Schadendorf, MD, PhD, University Hospital Essen, Essen, Germany, and German Cancer Consortium, Heidelberg, Germany; Mario Santinami, MD, Fondazione Istituto Nazionale Tumori, Milan, Italy; Victoria Atkinson, MD, Princess Alexandra Hospital, Gallipoli Medical
| | - Eduard Gasal
- Georgina V. Long, MD, PhD, Melanoma Institute Australia, The University of Sydney, and Royal North Shore and Mater Hospitals, Sydney, NSW, Australia; Reinhard Dummer, MD, University Hospital Zürich Skin Cancer Center, Zürich, Switzerland; Dirk Schadendorf, MD, PhD, University Hospital Essen, Essen, Germany, and German Cancer Consortium, Heidelberg, Germany; Mario Santinami, MD, Fondazione Istituto Nazionale Tumori, Milan, Italy; Victoria Atkinson, MD, Princess Alexandra Hospital, Gallipoli Medical
| | - Richard Kefford
- Georgina V. Long, MD, PhD, Melanoma Institute Australia, The University of Sydney, and Royal North Shore and Mater Hospitals, Sydney, NSW, Australia; Reinhard Dummer, MD, University Hospital Zürich Skin Cancer Center, Zürich, Switzerland; Dirk Schadendorf, MD, PhD, University Hospital Essen, Essen, Germany, and German Cancer Consortium, Heidelberg, Germany; Mario Santinami, MD, Fondazione Istituto Nazionale Tumori, Milan, Italy; Victoria Atkinson, MD, Princess Alexandra Hospital, Gallipoli Medical
| | - John M. Kirkwood
- Georgina V. Long, MD, PhD, Melanoma Institute Australia, The University of Sydney, and Royal North Shore and Mater Hospitals, Sydney, NSW, Australia; Reinhard Dummer, MD, University Hospital Zürich Skin Cancer Center, Zürich, Switzerland; Dirk Schadendorf, MD, PhD, University Hospital Essen, Essen, Germany, and German Cancer Consortium, Heidelberg, Germany; Mario Santinami, MD, Fondazione Istituto Nazionale Tumori, Milan, Italy; Victoria Atkinson, MD, Princess Alexandra Hospital, Gallipoli Medical
| | - Axel Hauschild
- Georgina V. Long, MD, PhD, Melanoma Institute Australia, The University of Sydney, and Royal North Shore and Mater Hospitals, Sydney, NSW, Australia; Reinhard Dummer, MD, University Hospital Zürich Skin Cancer Center, Zürich, Switzerland; Dirk Schadendorf, MD, PhD, University Hospital Essen, Essen, Germany, and German Cancer Consortium, Heidelberg, Germany; Mario Santinami, MD, Fondazione Istituto Nazionale Tumori, Milan, Italy; Victoria Atkinson, MD, Princess Alexandra Hospital, Gallipoli Medical
| |
Collapse
|
868
|
Yushak M, Mehnert J, Luke J, Poklepovic A. Approaches to High-Risk Resected Stage II and III Melanoma. Am Soc Clin Oncol Educ Book 2019; 39:e207-e211. [PMID: 31099653 DOI: 10.1200/edbk_239283] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Over the last decade, several therapies, including both targeted and immune checkpoint inhibitors, have dramatically changed the treatment landscape for patients with metastatic melanoma. These same therapies are now being used in the adjuvant setting with the hope of delaying or preventing the development of metastatic disease. Although phase III trials have shown a clear benefit for patients with resected bulky nodal disease, treatment decisions for patients with earlier-stage (high-risk stage II and stage IIIA) melanoma in the adjuvant setting are less straightforward given the small number of patients studied so far. Among patients with stage IIIB and worse disease, both targeted and immune checkpoint inhibitors have shown benefit in recurrence-free survival. Although a head-to-head comparison has not been completed, patient and tumor characteristics can guide the optimal treatment of an individual.
Collapse
Affiliation(s)
- Melinda Yushak
- 1 Winship Cancer Institute, Emory University, Atlanta, GA
| | - Janice Mehnert
- 2 Rutgers Cancer Institute of New Jersey, New Brunswick, NJ
| | - Jason Luke
- 3 University of Chicago Medicine, Chicago, IL
| | - Andrew Poklepovic
- 4 Massey Cancer Center, Virginia Commonwealth University, Richmond, VA
| |
Collapse
|
869
|
Affiliation(s)
- Robert Micieli
- Faculty of Medicine (Micieli, Pon), University of Toronto; Division of Dermatology, Sunnybrook Health Sciences Centre (Pon), Toronto, Ont
| | - Kucy Pon
- Faculty of Medicine (Micieli, Pon), University of Toronto; Division of Dermatology, Sunnybrook Health Sciences Centre (Pon), Toronto, Ont.
| |
Collapse
|
870
|
Ascierto PA, Bruzzi P, Eggermont A, Hamid O, Tawbi HA, van Akkooi A, Testori A, Caracò C, Puzanov I, Perrone F. The great debate at "Melanoma Bridge 2018", Naples, December 1st, 2018. J Transl Med 2019; 17:148. [PMID: 31077205 PMCID: PMC6509811 DOI: 10.1186/s12967-019-1892-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 04/25/2019] [Indexed: 12/19/2022] Open
Abstract
The great debate session at the 2018 Melanoma Bridge congress (November 29-December 1, Naples, Italy) featured counterpoint views from experts on three topical issues in melanoma. These were whether overall survival should still be the main endpoint for clinical trials in melanoma, whether anti-cytotoxic T-lymphocyte-associated antigen (CTLA)-4 is still the optimal choice of drug to use in combination with an anti-programmed death (PD)/PD-ligand (L)-1 agent, and the place of adjuvant versus neoadjuvant therapy in patients with melanoma. These three important debates are summarised in this report.
Collapse
Affiliation(s)
- Paolo A Ascierto
- Unit of Melanoma, Cancer Immunotherapy and Innovative Therapy, Istituto Nazionale Tumori IRCCS Fondazione "G. Pascale", Via Mariano Semmola, 80131, Naples, Italy.
| | - Paolo Bruzzi
- Clinical Epidemiology Unit, University Hospital "San Martino", Genoa, Italy
| | | | - Omid Hamid
- Clinical Research and Immunotherapy, The Angeles Clinic and Research Institute, Los Angeles, CA, USA
| | - Hussein A Tawbi
- Melanoma Clinical Research & Early Drug Development, Melanoma Medical Oncology, Investigational Cancer Therapeutics, UT MD Anderson Cancer Center, Houston, TX, USA
| | - Alexander van Akkooi
- Department of Surgical Oncology, Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital (NKI-AVL), Amsterdam, The Netherlands
| | | | - Corrado Caracò
- Department Melanoma, Soft Tissue, Muscle-Skeletal and Head-Neck, Istituto Nazionale Tumori IRCCS Fondazione "G. Pascale", Naples, Italy
| | - Igor Puzanov
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Francesco Perrone
- Clinical Trials Unit, Istituto Nazionale Tumori IRCCS Fondazione "G. Pascale", Naples, Italy
| |
Collapse
|
871
|
Luke JJ. Comprehensive Clinical Trial Data Summation for BRAF-MEK Inhibition and Checkpoint Immunotherapy in Metastatic Melanoma. Oncologist 2019; 24:e1197-e1211. [PMID: 31064886 PMCID: PMC6853121 DOI: 10.1634/theoncologist.2018-0876] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 03/22/2019] [Indexed: 12/30/2022] Open
Abstract
This review focuses on checkpoint and BRAF inhibitors, exploring outcomes based on clinical and disease characteristics to identify trends that might inform treatment decisions for the management of melanoma. Background. Immune checkpoint inhibitors, along with BRAF and MEK inhibitors, have dramatically changed the management of and outlook for patients with metastatic melanoma. Analyses of long‐term follow‐up data and subanalyses based on disease characteristics may inform clinical decision making. Methods. Reports of clinical trials in metastatic melanoma published between January 1, 2012, and August 30, 2018, were identified using PubMed (terms: melanoma AND [dabrafenib OR trametinib OR vemurafenib OR cobimetinib OR encorafenib OR ipilimumab OR nivolumab OR pembrolizumab]) and were systematically reviewed. Relevant congress proceedings were also assessed. Efficacy data from key phase III trials were analyzed and trends identified. Results. Substantial improvements in objective response rates, progression‐free survival, and overall survival were documented across 14 identified publications. Subgroup findings supported that patients with lower disease burden derive greater benefit than patients with more advanced disease, limiting the value of disease burden in the clinical decision‐making process. However, these agents consistently conferred benefits despite the presence of poor prognostic features. Several clinically relevant questions remain, including how best to sequence immune checkpoint inhibitors and combination targeted therapy. Conclusion. This research, coupled with ongoing investigations, including those on predictive biomarkers, suggests that the treatment decision‐making process is likely to become more nuanced. Implications for Practice. The management of melanoma has been rapidly advancing with new classes of agents, including immune checkpoint and BRAF inhibitors. With long‐term follow‐up, their impact on response rates and survival outcomes is well documented. Additional findings from subgroup analyses suggest that patients with lower disease burden derive greater benefit, yet both consistently confer benefit in patients with higher disease burden. Currently, there is a paucity of data to guide first‐line treatment selection between immunotherapy and BRAF‐targeted therapy in clinical practice or to estimate their impact when sequenced. Gaining these insights will facilitate a more nuanced management approach.
Collapse
Affiliation(s)
- Jason J Luke
- Department of Medicine, Division of Hematology/Oncology, University of Chicago Comprehensive Cancer Center, Chicago, Illinois, USA
| |
Collapse
|
872
|
Shader RI. Musings on Melanoma. Clin Ther 2019; 41:791-793. [DOI: 10.1016/j.clinthera.2019.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 04/05/2019] [Indexed: 11/30/2022]
|
873
|
Tan L, Sandhu S, Lee RJ, Li J, Callahan J, Ftouni S, Dhomen N, Middlehurst P, Wallace A, Raleigh J, Hatzimihalis A, Henderson MA, Shackleton M, Haydon A, Mar V, Gyorki DE, Oudit D, Dawson MA, Hicks RJ, Lorigan P, McArthur GA, Marais R, Wong SQ, Dawson SJ. Prediction and monitoring of relapse in stage III melanoma using circulating tumor DNA. Ann Oncol 2019; 30:804-814. [PMID: 30838379 PMCID: PMC6551451 DOI: 10.1093/annonc/mdz048] [Citation(s) in RCA: 127] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND The advent of effective adjuvant therapies for patients with resected melanoma has highlighted the need to stratify patients based on risk of relapse given the cost and toxicities associated with treatment. Here we assessed circulating tumor DNA (ctDNA) to predict and monitor relapse in resected stage III melanoma. PATIENTS AND METHODS Somatic mutations were identified in 99/133 (74%) patients through tumor tissue sequencing. Personalized droplet digital PCR (ddPCR) assays were used to detect known mutations in 315 prospectively collected plasma samples from mutation-positive patients. External validation was performed in a prospective independent cohort (n = 29). RESULTS ctDNA was detected in 37 of 99 (37%) individuals. In 81 patients who did not receive adjuvant therapy, 90% of patients with ctDNA detected at baseline and 100% of patients with ctDNA detected at the postoperative timepoint relapsed at a median follow up of 20 months. ctDNA detection predicted patients at high risk of relapse at baseline [relapse-free survival (RFS) hazard ratio (HR) 2.9; 95% confidence interval (CI) 1.5-5.6; P = 0.002] and postoperatively (HR 10; 95% CI 4.3-24; P < 0.001). ctDNA detection at baseline [HR 2.9; 95% CI 1.3-5.7; P = 0.003 and postoperatively (HR 11; 95% CI 4.3-27; P < 0.001] was also associated with inferior distant metastasis-free survival (DMFS). These findings were validated in the independent cohort. ctDNA detection remained an independent predictor of RFS and DMFS in multivariate analyses after adjustment for disease stage and BRAF mutation status. CONCLUSION Baseline and postoperative ctDNA detection in two independent prospective cohorts identified stage III melanoma patients at highest risk of relapse and has potential to inform adjuvant therapy decisions.
Collapse
Affiliation(s)
- L Tan
- Peter MacCallum Cancer Centre, Melbourne; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Australia
| | - S Sandhu
- Peter MacCallum Cancer Centre, Melbourne; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Australia
| | - R J Lee
- Molecular Oncology Group, Cancer Research UK Manchester Institute, Manchester; Faculty of Biology, Medicine and Health, The University of Manchester, Manchester
| | - J Li
- Peter MacCallum Cancer Centre, Melbourne; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Australia
| | - J Callahan
- Peter MacCallum Cancer Centre, Melbourne
| | - S Ftouni
- Peter MacCallum Cancer Centre, Melbourne
| | - N Dhomen
- Molecular Oncology Group, Cancer Research UK Manchester Institute, Manchester
| | - P Middlehurst
- Molecular Oncology Group, Cancer Research UK Manchester Institute, Manchester
| | - A Wallace
- Genomic Diagnostics Laboratory, Manchester Centre for Genomic Medicine, Manchester, UK
| | - J Raleigh
- Peter MacCallum Cancer Centre, Melbourne
| | | | - M A Henderson
- Peter MacCallum Cancer Centre, Melbourne; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Australia
| | | | | | - V Mar
- The Alfred Hospital, Melbourne
| | - D E Gyorki
- Peter MacCallum Cancer Centre, Melbourne; Department of Surgery, The University of Melbourne, Melbourne, Australia
| | - D Oudit
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester; The Christie NHS Foundation Trust, Manchester, UK
| | - M A Dawson
- Peter MacCallum Cancer Centre, Melbourne; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Australia; Centre for Cancer Research, The University of Melbourne, Melbourne, Australia
| | - R J Hicks
- Peter MacCallum Cancer Centre, Melbourne; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Australia
| | - P Lorigan
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester; The Christie NHS Foundation Trust, Manchester, UK
| | - G A McArthur
- Peter MacCallum Cancer Centre, Melbourne; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Australia
| | - R Marais
- Molecular Oncology Group, Cancer Research UK Manchester Institute, Manchester; Faculty of Biology, Medicine and Health, The University of Manchester, Manchester
| | - S Q Wong
- Peter MacCallum Cancer Centre, Melbourne
| | - S-J Dawson
- Peter MacCallum Cancer Centre, Melbourne; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Australia; Centre for Cancer Research, The University of Melbourne, Melbourne, Australia.
| |
Collapse
|
874
|
Keller J, Schwartz TL, Lizalek JM, Chang E, Patel AD, Hurley MY, Hsueh EC. Prospective validation of the prognostic 31-gene expression profiling test in primary cutaneous melanoma. Cancer Med 2019; 8:2205-2212. [PMID: 30950242 PMCID: PMC6536922 DOI: 10.1002/cam4.2128] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Revised: 02/27/2019] [Accepted: 03/13/2019] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Gene expression profiling (GEP) has been integrated into cancer treatment decision-making in multiple neoplasms. We prospectively evaluated the prognostic utility of the 31-GEP test (DecisionDx-Melanoma, Castle Biosciences, Inc) in cutaneous melanoma (CM) patients undergoing sentinel node biopsy (SNB). METHODS One hundred fifty-nine patients (age 26-88) diagnosed with melanoma between 01/2013 and 8/2015 underwent SNB and concurrent GEP testing. GEP results were reported as low-risk Class 1 (subclasses 1A and 1B) or high-risk Class 2 (subclasses 2A and 2B). Statistical analyses were performed with chi-square analysis, t tests, log-rank tests, and Cox proportional hazard models. Recurrence-free survival (RFS) and distant metastasis-free survival (DMFS) were estimated using Kaplan-Meier method. RESULTS Median follow-up was 44.9 months for event-free cases. Median Breslow thickness was 1.4 mm (0.2-15.0 mm). There were 117 Class 1 and 42 Class 2 patients. Gender, age, Breslow thickness, ulceration, SNB positivity, and AJCC stage were significantly associated with GEP classification (P < 0.05 for all). Recurrence and distant metastasis rates were 5% and 1% for Class 1 patients compared with 55% and 36% for Class 2 patients. Sensitivities of Class 2 and SNB for recurrence were 79% and 34%, respectively. Of 10 SNB-positive/Class 2 patients, 9 recurred. By multivariate analysis, only SNB result and GEP class were statistically associated with both RFS (P = 0.008 and 0.0001) and DMFS (P = 0.019 and 0.001). CONCLUSIONS Gene expression profiling Class 2 result and SNB positivity were independently associated with recurrence and distant metastasis in primary CM patients. GEP testing may have additive prognostic utility in initial staging work-up of these patients.
Collapse
Affiliation(s)
- Jennifer Keller
- Department of SurgerySaint Louis UniversitySt. LouisMissouri
| | | | | | - Ea‐sle Chang
- Department of SurgerySaint Louis UniversitySt. LouisMissouri
| | - Ashaki D. Patel
- Department of SurgerySaint Louis UniversitySt. LouisMissouri
| | - Maria Y. Hurley
- Department of DermatologySaint Louis UniversitySt. LouisMissouri
| | - Eddy C. Hsueh
- Department of SurgerySaint Louis UniversitySt. LouisMissouri
| |
Collapse
|
875
|
Lee JH, Saw RP, Thompson JF, Lo S, Spillane AJ, Shannon KF, Stretch JR, Howle J, Menzies AM, Carlino MS, Kefford RF, Long GV, Scolyer RA, Rizos H. Pre-operative ctDNA predicts survival in high-risk stage III cutaneous melanoma patients. Ann Oncol 2019; 30:815-822. [PMID: 30860590 PMCID: PMC6551453 DOI: 10.1093/annonc/mdz075] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND The outcomes of patients with stage III cutaneous melanoma who undergo complete surgical resection can be highly variable, and estimation of individual risk of disease recurrence and mortality remains imprecise. With recent demonstrations of effective adjuvant targeted and immune checkpoint inhibitor therapy, more precise stratification of patients for costly and potentially toxic adjuvant therapy is needed. We report the utility of pre-operative circulating tumour DNA (ctDNA) in patients with high-risk stage III melanoma. PATIENTS AND METHODS ctDNA was analysed in blood specimens that were collected pre-operatively from 174 patients with stage III melanoma undergoing complete lymph node (LN) dissection. Cox regression analyses were used to evaluate the prognostic significance of ctDNA for distant metastasis recurrence-free survival and melanoma-specific survival (MSS). RESULTS The detection of ctDNA in the discovery and validation cohort was 34% and 33%, respectively, and was associated with larger nodal melanoma deposit, higher number of melanoma involved LNs, more advanced stage and high lactate dehydrogenase (LDH) levels. Detectable ctDNA was significantly associated with worse MSS in the discovery [hazard ratio (HR) 2.11 P < 0.01] and validation cohort (HR 2.29, P = 0.04) and remained significant in a multivariable analysis (HR 1.85, P = 0.04). ctDNA further sub-stratified patients with AJCC stage III substage, with increasing significance observed in more advanced stage melanoma. CONCLUSION Pre-operative ctDNA predicts MSS in high-risk stage III melanoma patients undergoing complete LN dissection, independent of stage III substage. This biomarker may have an important role in determining prognosis and stratifying patients for adjuvant treatment.
Collapse
Affiliation(s)
- J H Lee
- Faculty of Medicine and Health Sciences, Macquarie University, Macquarie Park, NSW; Melanoma Institute Australia, Wollstonecraft, NSW
| | - R P Saw
- Melanoma Institute Australia, Wollstonecraft, NSW; Department of Melanoma and Surgical Oncology, Royal Prince Alfred Hospital, Camperdown, NSW; Sydney Medical School, The University of Sydney, Camperdown, NSW
| | - J F Thompson
- Melanoma Institute Australia, Wollstonecraft, NSW; Department of Melanoma and Surgical Oncology, Royal Prince Alfred Hospital, Camperdown, NSW; Sydney Medical School, The University of Sydney, Camperdown, NSW
| | - S Lo
- Melanoma Institute Australia, Wollstonecraft, NSW; Sydney Medical School, The University of Sydney, Camperdown, NSW
| | - A J Spillane
- Melanoma Institute Australia, Wollstonecraft, NSW; Northern Sydney Cancer Centre, Royal North Shore Hospital, St Leonards, NSW
| | - K F Shannon
- Melanoma Institute Australia, Wollstonecraft, NSW; Chris O'Brien Lifehouse, Camperdown, NSW
| | - J R Stretch
- Melanoma Institute Australia, Wollstonecraft, NSW
| | - J Howle
- Crown Princess Mary Cancer Centre, Westmead and Blacktown hospitals, Wentworthville, NSW
| | - A M Menzies
- Melanoma Institute Australia, Wollstonecraft, NSW; Sydney Medical School, The University of Sydney, Camperdown, NSW; Northern Sydney Cancer Centre, Royal North Shore Hospital, St Leonards, NSW
| | - M S Carlino
- Melanoma Institute Australia, Wollstonecraft, NSW; Sydney Medical School, The University of Sydney, Camperdown, NSW; Crown Princess Mary Cancer Centre, Westmead and Blacktown hospitals, Wentworthville, NSW
| | - R F Kefford
- Faculty of Medicine and Health Sciences, Macquarie University, Macquarie Park, NSW; Melanoma Institute Australia, Wollstonecraft, NSW; Crown Princess Mary Cancer Centre, Westmead and Blacktown hospitals, Wentworthville, NSW
| | - G V Long
- Melanoma Institute Australia, Wollstonecraft, NSW; Sydney Medical School, The University of Sydney, Camperdown, NSW; Northern Sydney Cancer Centre, Royal North Shore Hospital, St Leonards, NSW
| | - R A Scolyer
- Melanoma Institute Australia, Wollstonecraft, NSW; Sydney Medical School, The University of Sydney, Camperdown, NSW; Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
| | - H Rizos
- Faculty of Medicine and Health Sciences, Macquarie University, Macquarie Park, NSW; Melanoma Institute Australia, Wollstonecraft, NSW.
| |
Collapse
|
876
|
Falk Delgado A, Zommorodi S, Falk Delgado A. Sentinel Lymph Node Biopsy and Complete Lymph Node Dissection for Melanoma. Curr Oncol Rep 2019; 21:54. [PMID: 31028497 PMCID: PMC6486528 DOI: 10.1007/s11912-019-0798-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
PURPOSE OF REVIEW The main surgical treatment for invasive malignant melanoma consists of wide surgical and examination of the sentinel node and in selected cases complete lymph node dissection. The aim of this review is to present data for the optimal surgical management of patients with malignant melanoma. RECENT FINDINGS A surgical excision margin of 1-2 cm is recommended for invasive melanoma depending on the thickness of the melanoma. Sentinel node biopsy may be considered for patients with at least T1b melanomas thickness 0.8 to 1.0 mm or less than 0.8 mm Breslow thickness with ulceration, classified as T1b lesion, per recent AJCC guidelines. Two randomized controlled trials have been published-DeCOG (German Dermatologic Cooperative Oncology Group Selective Lymphadenectomy) and MSLT-2 (Multicenter Selective Lymphadenectomy Trial) comparing the complete lymph node dissection (CLND) with observation after positive sentinel node biopsy. In the MSLT-2 study, the disease control rate was improved in the immediate CLND group compared with observation but there was no difference in 3-year melanoma specific survival (86% ± 1.3% and 86% ± 1.2%, respectively; p = 0.42). Isolated limb perfusion (ILP) or isolated limb infusion (ILI) with melphalan and actinomycin D is recommended for large and multiple in-transit metastases and satellite metastases in the extremities when local excision is considered ineffective or too extensive. In light of new adjuvant treatment options and new indications for checkpoint inhibitors, and the lack of survival benefit after CLND, we can expect open surgery to decrease in melanoma disease.
Collapse
Affiliation(s)
- Alberto Falk Delgado
- Department of Plastic Surgery, Uppsala University, Ing 85, Akademiska Sjukhuset, 75185, Uppsala, Sweden.
| | - Sayid Zommorodi
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Department of Plastic Surgery, Karolinska University Hospital, Stockholm, Sweden
| | - Anna Falk Delgado
- Clinical neurosciences, Karolinska Institutet, Stockholm, Sweden
- Department of Neuroradiology, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
877
|
Kint N, Vlayen S, Delforge M. The treatment of multiple myeloma in an era of precision medicine. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2019. [DOI: 10.1080/23808993.2019.1606672] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Affiliation(s)
- Nicolas Kint
- Department of Internal Medicine, Hematology, University Hospitals Leuven, Leuven, Belgium
- Stem Cell Institute Leuven, KU Leuven, Leuven, Belgium
| | - Sophie Vlayen
- Department of Internal Medicine, Hematology, University Hospitals Leuven, Leuven, Belgium
- Stem Cell Institute Leuven, KU Leuven, Leuven, Belgium
| | - Michel Delforge
- Department of Internal Medicine, Hematology, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
878
|
Broman KK, Dossett LA, Sun J, Eroglu Z, Zager JS. Update on BRAF and MEK inhibition for treatment of melanoma in metastatic, unresectable, and adjuvant settings. Expert Opin Drug Saf 2019; 18:381-392. [PMID: 30977681 DOI: 10.1080/14740338.2019.1607289] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Selective inhibition of the MAPK pathway with BRAF and MEK inhibitors has emerged as a key component of the treatment of BRAF-mutant unresectable/locally advanced metastatic melanoma. AREAS COVERED Current data are presented on the efficacy and safety of BRAFi + MEKi combination therapy (dabrafenib/trametinib, vemurafenib/cobimetinib, and encorafenib/binimetinib) from phase I, II, and III trials in the unresectable/locally advanced metastatic setting, as well as neoadjuvant and adjuvant applications. The theoretical basis, pre-clinical findings, clinical trial results and current ongoing clinical studies of combined BRAF/MEK inhibition with immunotherapy, also known as 'triplet therapy,' are also explored. EXPERT OPINION Combination therapy with BRAF and MEK inhibitors dramatically improves response rates, progression-free survival and overall survival in patients with BRAF-mutant metastatic melanoma compared to historical treatments such as chemotherapy. Some serious adverse effects, including cutaneous squamous cell carcinoma, are attenuated with combination therapy, while less severe and reversible effects including pyrexia, left ventricular dysfunction, and ocular events can be more common with combination therapy. Existing data are insufficient to recommend triplet therapy, or a particular treatment sequence, with respect to BRAF and MEK inhibitors and immune therapies, though results from multiple ongoing trials are anticipated.
Collapse
Affiliation(s)
| | - Lesly A Dossett
- b Division of Surgical Oncology , University of Michigan , Ann Arbor , MI , USA
| | - James Sun
- a Department of Cutaneous Oncology , Moffitt Cancer Center , Tampa , FL , USA
| | - Zeynep Eroglu
- a Department of Cutaneous Oncology , Moffitt Cancer Center , Tampa , FL , USA
| | - Jonathan S Zager
- a Department of Cutaneous Oncology , Moffitt Cancer Center , Tampa , FL , USA
| |
Collapse
|
879
|
Abstract
PURPOSE OF REVIEW We review the results from relevant clinical trials and discuss current strategies in the melanoma adjuvant setting. RECENT FINDINGS The favorable therapeutic efficacy and the significant less toxicity of nivolumab compared with ipilimumab, fully substitutes today's approval of ipilimumab, regardless mutation status, whereas in BRAF-mutated patients, dabrafenib and trametinib seem to confirm their high efficacy also in adjuvant setting. The use of interferon is restricted to patients with ulcerated melanoma and countries with no access to the new drugs. SUMMARY Systemic adjuvant treatment after complete disease resection in high-risk melanoma patients aims to increase relapse-free survival (RFS) and overall survival (OS). According to the eighth edition of melanoma classification of American Joint Committee on Cancer (AJCC), the prognosis in stage III patients is heterogeneous and depends not only on N (nodal) but also on T (tumor thickness) category criteria. Recent data from randomized, phase-3 clinical trials analyzing the use of adjuvant anti-programmed death-1 and targeted therapies ultimately affect the standard of care and change the landscape of the adjuvant treatment.
Collapse
|
880
|
Banting S, Milne D, Thorpe T, Na L, Spillane J, Speakman D, Henderson MA, Gyorki DE. Negative Sentinel Lymph Node Biopsy in Patients with Melanoma: The Patient's Perspective. Ann Surg Oncol 2019; 26:2263-2267. [PMID: 31011899 DOI: 10.1245/s10434-019-07375-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Indexed: 11/18/2022]
Abstract
BACKGROUND The majority of patients undergoing sentinel lymph node biopsy (SLNB) for melanoma will have a negative SLN. The long-term sequelae of a negative result are important when discussing this staging investigation with patients. The objective of this study was to assess rates of lymphoedema and quality of life for these patients. METHODS A prospective, cross-sectional study was performed on patients under routine follow-up with a history of melanoma, who had undergone sentinel lymph node biopsy where no metastasis was found (N0) at a high-volume melanoma centre. Relevant limbs were measured to assess for lymphoedema and patients completed the FACT-M quality of life instrument and a study specific questionnaire. RESULTS A total of 102 patients were recruited. Wound complications were observed in 25% and lymphoedema in 2% of patients. Physical and functional well-being scores were lowest in patients seen within 3 months of their SLNB. Functional well-being and quality of life improved over the 2 years following the procedure. CONCLUSIONS SLNB has low complication rates. The procedure is associated with a short-term impact on patient quality of life and well-being. The vast majority of patients are pleased with the outcomes of this procedure and the information that it provides.
Collapse
Affiliation(s)
- Sarah Banting
- Department of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Melbourne Medical School, University of Melbourne, Melbourne, Australia
| | - Donna Milne
- Department of Cancer Experiences Research, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Tina Thorpe
- Department of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Lumine Na
- Centre for Biostatistics and Clinical Trials, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - John Spillane
- Department of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - David Speakman
- Department of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Michael A Henderson
- Department of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,Department of Surgery, University of Melbourne, Melbourne, Australia
| | - David E Gyorki
- Department of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia. .,Department of Surgery, University of Melbourne, Melbourne, Australia.
| |
Collapse
|
881
|
Fujisawa Y, Yoshikawa S, Minagawa A, Takenouchi T, Yokota K, Uchi H, Noma N, Nakamura Y, Asai J, Kato J, Fujiwara S, Fukushima S, Uehara J, Hoashi T, Kaji T, Fujimura T, Namikawa K, Yoshioka M, Murata N, Ogata D, Matsuyama K, Hatta N, Shibayama Y, Fujiyama T, Ishikawa M, Yamada D, Kishi A, Nakamura Y, Shimiauchi T, Fujii K, Fujimoto M, Ihn H, Katoh N. Classification of 3097 patients from the Japanese melanoma study database using the American joint committee on cancer eighth edition cancer staging system. J Dermatol Sci 2019; 94:284-289. [PMID: 31023613 DOI: 10.1016/j.jdermsci.2019.04.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 04/10/2019] [Accepted: 04/11/2019] [Indexed: 10/27/2022]
Abstract
BACKGROUND The American Joint Committee on Cancer (AJCC) 8th Edition Cancer Staging System was implemented in 2018; however, it has not been validated in an Asian melanoma population. OBJECTIVE The purpose of this study was to validate the new system using a cohort of Japanese melanoma patients. METHODS The AJCC 7th and 8th Editions were used for TNM classification of patients in a database established by the Japanese Melanoma Study Group. Patient data with sufficient information to be applicable to the AJCC 8th staging were selected. The Kaplan-Meier method was used to estimate disease-specific survival and relapse-free survival. RESULTS In total, data for 3097 patients were analyzed. The 5-year disease-specific survival according to the 7th and 8th Edition staging system were as follows: IA = 98.5%/97.9%; IB = 95.4%/96.2%; IIA = 94.2%/94.1%; IIB = 84.6%/84.4%; IIC = 72.2%/72.2%; IIIA = 76.2%/87.5%; IIIB = 60.7%/72.6%; IIIC = 42.0%/55.3% and IIID = none/26.0%. The 5-year relapse-free survival according to the 7th and 8th Edition staging was as follows: IA = 94.5%/92.7%; IB = 85.4%/85.3%; IIA = 80.1%/79.4%; IIB = 71.4%/70.6%; IIC = 56.8%/55.7%; IIIA = 56.8%/69.4%; IIIB = 42.6%/56.8%; IIIC = 20.0%/33.3% and IIID = none/6.5%. CONCLUSION The results show that new staging system could efficiently classify our Japanese melanoma cohort. Although there was no difference in Stage I and II disease between the 7th and 8th Edition systems, we should be careful in managing Stage III disease since the survival curves of the 8th Edition staging were completely different from the 7th Edition. Moreover, our results indicate that adjuvant therapies for Stage IIB and IIC should be developed, since the relapse-free survival for these stages were equivalent to Stage IIIA and IIIB, respectively.
Collapse
Affiliation(s)
- Yasuhiro Fujisawa
- Japanese Melanoma Study Group, Japan; Prognosis and Statistical Investigation Committee of the Japanese Skin Cancer Society, Japan; Department of Dermatology, University of Tsukuba, Japan.
| | - Shusuke Yoshikawa
- Japanese Melanoma Study Group, Japan; Department of Dermatology, Shizuoka Cancer Center, Japan
| | - Akane Minagawa
- Japanese Melanoma Study Group, Japan; Department of Dermatology, Shinshu University School of Medicine, Japan
| | - Tatsuya Takenouchi
- Japanese Melanoma Study Group, Japan; Department of Dermatology, Niigata Cancer Center, Japan
| | - Kenji Yokota
- Japanese Melanoma Study Group, Japan; Department of Dermatology, University of Nagoya, Japan
| | - Hiroshi Uchi
- Japanese Melanoma Study Group, Japan; Department of Dermatology, University of Kyushu, Japan
| | - Naoki Noma
- Japanese Melanoma Study Group, Japan; Department of Dermatology, Osaka City University, Japan
| | - Yasuhiro Nakamura
- Japanese Melanoma Study Group, Japan; Department of Dermatology, Saitama Medical University International Medical Center, Japan
| | - Jun Asai
- Japanese Melanoma Study Group, Japan; Department of Dermatology, Kyoto Prefectural University of Medicine, Japan
| | - Junji Kato
- Japanese Melanoma Study Group, Japan; Department of Dermatology, Sapporo Medical University, Japan
| | - Susumu Fujiwara
- Japanese Melanoma Study Group, Japan; Department of Dermatology, Kobe University, Japan
| | - Satoshi Fukushima
- Japanese Melanoma Study Group, Japan; Department of Department of Dermatology and Plastic Surgery, Kumamoto University, Japan
| | - Jiro Uehara
- Japanese Melanoma Study Group, Japan; Department of Dermatology, Asahikawa Medical University, Japan
| | - Toshihiko Hoashi
- Japanese Melanoma Study Group, Japan; Department of Dermatology, Nippon Medical School, Japan
| | - Tatsuya Kaji
- Japanese Melanoma Study Group, Japan; Department of Dermatology, Okayama University Graduate School of Medicine, Japan
| | - Taku Fujimura
- Japanese Melanoma Study Group, Japan; Department of Dermatology, Tohoku University Graduate School of Medicine, Japan
| | - Kenjiro Namikawa
- Japanese Melanoma Study Group, Japan; Department of Dermatologic Oncology, National Cancer Center Hospital, Japan
| | - Manabu Yoshioka
- Japanese Melanoma Study Group, Japan; Department of Dermatology, University of Occupational Environment Health, Japan
| | - Naoki Murata
- Japanese Melanoma Study Group, Japan; Department of Plastic Surgery, University of Hokkaido, Japan
| | - Dai Ogata
- Japanese Melanoma Study Group, Japan; Department of Dermatology, Saitama Medical University, Japan
| | - Kanako Matsuyama
- Japanese Melanoma Study Group, Japan; Department of Dermatology, University of Gifu, Japan
| | - Naohito Hatta
- Japanese Melanoma Study Group, Japan; Department of Dermatology, Toyama Prefectural Central Hospital, Japan
| | - Yoshitsugu Shibayama
- Japanese Melanoma Study Group, Japan; Department of Dermatology, Fukuoka University, Japan
| | - Toshiharu Fujiyama
- Japanese Melanoma Study Group, Japan; Department of Dermatology, Hamamatsu University School of Medicine, Japan
| | - Masashi Ishikawa
- Japanese Melanoma Study Group, Japan; Department of Dermatology, Saitama Prefectural Cancer Center, Japan
| | - Daisuke Yamada
- Japanese Melanoma Study Group, Japan; Department of Dermatology, University of Tokyo, Japan
| | - Akiko Kishi
- Japanese Melanoma Study Group, Japan; Department of Dermatology, Toranomon Hospital, Japan
| | | | - Takatoshi Shimiauchi
- Prognosis and Statistical Investigation Committee of the Japanese Skin Cancer Society, Japan; Department of Dermatology, Hamamatsu University School of Medicine, Japan
| | - Kazuyasu Fujii
- Prognosis and Statistical Investigation Committee of the Japanese Skin Cancer Society, Japan; Department of Dermatology, Kagoshima University, Japan
| | | | - Hironobu Ihn
- Prognosis and Statistical Investigation Committee of the Japanese Skin Cancer Society, Japan; Department of Department of Dermatology and Plastic Surgery, Kumamoto University, Japan
| | - Norito Katoh
- Prognosis and Statistical Investigation Committee of the Japanese Skin Cancer Society, Japan; Department of Dermatology, Kyoto Prefectural University of Medicine, Japan
| |
Collapse
|
882
|
Cohen JV, Sullivan RJ. Developments in the Space of New MAPK Pathway Inhibitors for BRAF-Mutant Melanoma. Clin Cancer Res 2019; 25:5735-5742. [PMID: 30992297 DOI: 10.1158/1078-0432.ccr-18-0836] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 01/28/2019] [Accepted: 04/12/2019] [Indexed: 12/19/2022]
Abstract
The characterization of the MAPK signaling pathway has led to the development of multiple promising targeted therapy options for a subset of patients with metastatic melanoma. The combination of BRAF and MEK inhibitors represents an FDA-approved standard of care in patients with metastatic and resected BRAF-mutated melanoma. There are currently three FDA-approved BRAF/MEK inhibitor combinations for the treatment of patients with BRAF-mutated melanoma. Although there have been significant advances in the field of targeted therapy, further exploration of new targets within the MAPK pathway will strengthen therapeutic options for patients. Important clinical and translational research focuses on mechanisms of resistance, predictive biomarkers, and challenging patient populations such as those with brain metastases or resected melanoma.
Collapse
Affiliation(s)
- Justine V Cohen
- Division of Medical Oncology, Department of Medicine, Massachusetts General Hospital Cancer Center, Center for Melanoma, Harvard Medical School, Boston, Massachusetts
| | - Ryan J Sullivan
- Division of Medical Oncology, Department of Medicine, Massachusetts General Hospital Cancer Center, Center for Melanoma, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
883
|
Gartrell RD, Marks DK, Rizk EM, Bogardus M, Gérard CL, Barker LW, Fu Y, Esancy CL, Li G, Ji J, Rui S, Ernstoff MS, Taback B, Pabla S, Chang R, Lee SJ, Krolewski JJ, Morrison C, Horst BA, Saenger YM. Validation of Melanoma Immune Profile (MIP), a Prognostic Immune Gene Prediction Score for Stage II-III Melanoma. Clin Cancer Res 2019; 25:2494-2502. [PMID: 30647081 PMCID: PMC6594682 DOI: 10.1158/1078-0432.ccr-18-2847] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 11/14/2018] [Accepted: 01/11/2019] [Indexed: 01/17/2023]
Abstract
PURPOSE Biomarkers are needed to stratify patients with stage II-III melanoma for clinical trials of adjuvant therapy because, while immunotherapy is protective, it also confers the risk of severe toxicity. We previously defined and validated a 53-immune gene melanoma immune profile (MIP) predictive both of distant metastatic recurrence and of disease-specific survival (DSS). Here, we test MIP on a third independent population. EXPERIMENTAL DESIGN A retrospective cohort of 78 patients with stage II-III primary melanoma was analyzed using the NanoString assay to measure expression of 53 target genes, and MIP score was calculated. Statistical analysis correlating MIP with DSS, overall survival, distant metastatic recurrence, and distant metastasis-free interval was performed using ROC curves, Kaplan-Meier curves, and standard univariable and multivariable Cox proportional hazards models. RESULTS MIP significantly distinguished patients with distant metastatic recurrence from those without distant metastatic recurrence using ROC curve analysis (AUC = 0.695; P = 0.008). We defined high- and low-risk groups based on the cutoff defined by this ROC curve and find that MIP correlates with both DSS and overall survival by ROC curve analysis (AUC = 0.719; P = 0.004 and AUC = 0.698; P = 0.004, respectively). Univariable Cox regression reveals that a high-risk MIP score correlates with DSS (P = 0.015; HR = 3.2). CONCLUSIONS MIP identifies patients with low risk of death from melanoma and may constitute a clinical tool to stratify patients with stage II-III melanoma for enrollment in clinical trials.
Collapse
Affiliation(s)
| | - Douglas K Marks
- Columbia University Irving Medical Center, New York, New York
| | | | - Margaret Bogardus
- College of Physician and Surgeons, Columbia University, New York, New York
| | | | - Luke W Barker
- College of Physician and Surgeons, Columbia University, New York, New York
| | - Yichun Fu
- College of Physician and Surgeons, Columbia University, New York, New York
| | - Camden L Esancy
- Columbia University Irving Medical Center, New York, New York
| | - Gen Li
- Mailman School of Public Health, Columbia University, New York, New York
| | - Jiayi Ji
- Mailman School of Public Health, Columbia University, New York, New York
| | - Shumin Rui
- Mailman School of Public Health, Columbia University, New York, New York
| | | | - Bret Taback
- Columbia University Irving Medical Center, New York, New York
| | | | - Rui Chang
- University of Arizona, Tucson, Arizona
| | - Sandra J Lee
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | | | | | | | | |
Collapse
|
884
|
Belgrano V, Mattsson J, Nilsson J, Olofsson Bagge R, Katsarelias D. BRAF status as a predictive factor for response in isolated limb perfusion. Int J Hyperthermia 2019; 36:511-515. [DOI: 10.1080/02656736.2019.1601778] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Affiliation(s)
- Valerio Belgrano
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Sahlgrenska Cancer Center, University of Gothenburg, Gothenburg, Sweden
| | - Jan Mattsson
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Department of Surgery, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Jonas Nilsson
- Sahlgrenska Cancer Center, University of Gothenburg, Gothenburg, Sweden
| | - Roger Olofsson Bagge
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Sahlgrenska Cancer Center, University of Gothenburg, Gothenburg, Sweden
- Department of Surgery, Sahlgrenska University Hospital, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Dimitrios Katsarelias
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Department of Surgery, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
885
|
Laurent M, Brahmi M, Dufresne A, Meeus P, Karanian M, Ray-Coquard I, Blay JY. Adjuvant therapy with imatinib in gastrointestinal stromal tumors (GISTs)-review and perspectives. Transl Gastroenterol Hepatol 2019; 4:24. [PMID: 31143845 DOI: 10.21037/tgh.2019.03.07] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 03/19/2019] [Indexed: 12/21/2022] Open
Abstract
Gastrointestinal stromal tumors (GISTs) are the most common mesenchymal tumor of the gastrointestinal (GI) tract. Most of them (>75%) are driven by an oncogenic initiating event involving activating mutations of the genes encoding for tyrosine kinase, KIT or platelet derived growth factor receptor alpha (PDGFRA). Efficacy of the tyrosine kinase Inhibitor imatinib is now well established for advanced disease. For localized GISTs, 3 years treatment is the recommended adjuvant therapy for high risk patients. Whether a longer duration and selection of patients for this adjuvant therapy in localized GISTs remains is not yet established (PERSIST-5 study). Similarly, it will be important to further refine the definition of the population of GIST patients at high risk of relapse including molecular criteria (Annals of Oncology, ESMO guidelines 2018). This review aims to describe current knowledges on the issue of adjuvant therapy of primary GISTs in view of available results.
Collapse
Affiliation(s)
- Marie Laurent
- Department of Medical Oncology, Centre Leon Berard, Lyon, France
| | - Mehdi Brahmi
- Department of Medical Oncology, Centre Leon Berard, Lyon, France
| | - Armelle Dufresne
- Department of Medical Oncology, Centre Leon Berard, Lyon, France
| | - Pierre Meeus
- Department of Medical Oncology, Centre Leon Berard, Lyon, France
| | - Marie Karanian
- Department of Medical Oncology, Centre Leon Berard, Lyon, France
| | | | - Jean-Yves Blay
- Department of Medical Oncology, Centre Leon Berard, Lyon, France
| |
Collapse
|
886
|
Eggermont A, Suciu S, Kicinski M. Estimation of Distant Metastasis-free Survival in Trials of Adjuvant Therapy for Melanoma. N Engl J Med 2019; 380:1376-1377. [PMID: 30943345 DOI: 10.1056/nejmc1902228] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
887
|
Rotman J, Koster BD, Jordanova ES, Heeren AM, de Gruijl TD. Unlocking the therapeutic potential of primary tumor-draining lymph nodes. Cancer Immunol Immunother 2019; 68:1681-1688. [PMID: 30944963 PMCID: PMC6805797 DOI: 10.1007/s00262-019-02330-y] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 03/22/2019] [Indexed: 01/24/2023]
Abstract
Lymph nodes draining the primary tumor are essential for the initiation of an effective anti-tumor T-cell immune response. However, cancer-derived immune suppressive factors render the tumor-draining lymph nodes (TDLN) immune compromised, enabling tumors to invade and metastasize. Unraveling the different mechanisms underlying this immune escape will inform therapeutic intervention strategies to halt tumor spread in early clinical stages. Here, we review our findings from translational studies in melanoma, breast, and cervical cancer and discuss clinical opportunities for local immune modulation of TDLN in each of these indications.
Collapse
Affiliation(s)
- Jossie Rotman
- Department of Obstetrics and Gynecology, Center for Gynecological Oncology Amsterdam (CGOA), Amsterdam UMC, Cancer Center Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Bas D Koster
- Department of Medical Oncology, Amsterdam UMC, Cancer Center Amsterdam, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Ekaterina S Jordanova
- Department of Obstetrics and Gynecology, Center for Gynecological Oncology Amsterdam (CGOA), Amsterdam UMC, Cancer Center Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - A Marijne Heeren
- Department of Medical Oncology, Amsterdam UMC, Cancer Center Amsterdam, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Tanja D de Gruijl
- Department of Medical Oncology, Amsterdam UMC, Cancer Center Amsterdam, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands.
| |
Collapse
|
888
|
Smithers BM. Conclusions from quality of life studies in patients with resected high-risk melanoma: one part of the full story. Lancet Oncol 2019; 20:611-612. [PMID: 30928621 DOI: 10.1016/s1470-2045(19)30176-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 02/19/2019] [Indexed: 10/27/2022]
Affiliation(s)
- B Mark Smithers
- Queensland Melanoma Project, University of Queensland, Princess Alexandra Hospital, Ipswich Road, Brisbane 4102, QLD, Australia.
| |
Collapse
|
889
|
Many ways to resistance: How melanoma cells evade targeted therapies. Biochim Biophys Acta Rev Cancer 2019; 1871:313-322. [DOI: 10.1016/j.bbcan.2019.02.002] [Citation(s) in RCA: 117] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 01/20/2019] [Accepted: 02/13/2019] [Indexed: 12/25/2022]
|
890
|
Wuthrick EJ, Chablani P. Do Not Forget About the Importance of Loco-Regional Therapy in Melanoma Management. Semin Radiat Oncol 2019; 29:166-170. [PMID: 30827455 DOI: 10.1016/j.semradonc.2018.11.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Immunotherapy agents have significantly changed the landscape of melanoma treatment over the past decade. Paradigm shifts in treatment require reanalysis of the treatment algorithms in melanoma. Despite surgical excision, certain high risk patients with desmoplastic melanoma remain at high risk for local recurrence and retrospective data suggests improvement in local control with adjuvant radiation therapy. Likewise, despite surgical excision and effective systemic therapy agents, patients with extracapsular extension and other high risk features are at substantial risk of nodal basin (regional) recurrence. Adjuvant radiation therapy has been demonstrated to reduce the local recurrence risk. Despite these benefits, adjuvant radiation therapy in melanoma remains controversial in part because its use has not been definitively demonstrated to improve overall or disease-free survival in a randomized prospective study.
Collapse
Affiliation(s)
- Evan J Wuthrick
- Department of Radiation Oncology, Moffitt Cancer Center, Tampa, FL.
| | | |
Collapse
|
891
|
Morgado-Carrasco D, Terc F, Ertekin S, Ferrandiz L. Good News on Adjuvant Therapy for Advanced Cutaneous Melanoma. ACTAS DERMO-SIFILIOGRAFICAS 2019. [DOI: 10.1016/j.adengl.2017.10.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
|
892
|
Vetto JT, Hsueh EC, Gastman BR, Dillon LD, Monzon FA, Cook RW, Keller J, Huang X, Fleming A, Hewgley P, Gerami P, Leachman S, Wayne JD, Berger AC, Fleming MD. Guidance of sentinel lymph node biopsy decisions in patients with T1–T2 melanoma using gene expression profiling. Future Oncol 2019; 15:1207-1217. [DOI: 10.2217/fon-2018-0912] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Aim: Can gene expression profiling be used to identify patients with T1–T2 melanoma at low risk for sentinel lymph node (SLN) positivity? Patients & methods: Bioinformatics modeling determined a population in which a 31-gene expression profile test predicted <5% SLN positivity. Multicenter, prospectively-tested (n = 1421) and retrospective (n = 690) cohorts were used for validation and outcomes, respectively. Results: Patients 55–64 years and ≥65 years with a class 1A (low-risk) profile had SLN positivity rates of 4.9% and 1.6%. Class 2B (high-risk) patients had SLN positivity rates of 30.8% and 11.9%. Melanoma-specific survival was 99.3% for patients ≥55 years with class 1A, T1–T2 tumors and 55.0% for class 2B, SLN-positive, T1–T2 tumors. Conclusion: The 31-gene expression profile test identifies patients who could potentially avoid SLN biopsy.
Collapse
Affiliation(s)
- John T Vetto
- Division of Surgical Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Eddy C Hsueh
- Department of Surgery, St Louis University, St Louis, MO 63110, USA
| | - Brian R Gastman
- Department of Plastic Surgery, Cleveland Clinic Lerner Research Institute, Cleveland, OH 44915, USA
| | - Larry D Dillon
- Larry D Dillon Surgical Oncology & General Surgery, Colorado Springs, CO 80907, USA
| | | | - Robert W Cook
- Castle Biosciences, Inc., Friendswood, TX 77546, USA
| | - Jennifer Keller
- Department of Surgery, St Louis University, St Louis, MO 63110, USA
| | - Xin Huang
- Division of Surgical Oncology, Department of Surgery, The University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Andrew Fleming
- Division of Surgical Oncology, Department of Surgery, The University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Preston Hewgley
- Division of Surgical Oncology, Department of Surgery, The University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Pedram Gerami
- Department of Dermatology, Northwestern University Feinberg School of Medicine, Chicago IL 60611, USA
- Skin Cancer Institute, Northwestern University, Lurie Comprehensive Cancer Center, Chicago, IL 60611, USA
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago IL 60611, USA
| | - Sancy Leachman
- Department of Dermatology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Jeffrey D Wayne
- Department of Dermatology, Northwestern University Feinberg School of Medicine, Chicago IL 60611, USA
- Skin Cancer Institute, Northwestern University, Lurie Comprehensive Cancer Center, Chicago, IL 60611, USA
- Department of Surgical Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Adam C Berger
- Department of Surgery, Thomas Jefferson University Hospital, Philadelphia, PA 19017, USA
| | - Martin D Fleming
- Division of Surgical Oncology, Department of Surgery, The University of Tennessee Health Science Center, Memphis, TN 38163, USA
| |
Collapse
|
893
|
Suarez-Kelly LP, Levine KM, Olencki TE, Del Campo SEM, Streacker EA, Brooks TR, Karpa VI, Markowitz J, Bingman AK, Geyer SM, Kendra KL, Carson WE. A pilot study of interferon-alpha-2b dose reduction in the adjuvant therapy of high-risk melanoma. Cancer Immunol Immunother 2019; 68:619-629. [PMID: 30725205 PMCID: PMC6447692 DOI: 10.1007/s00262-019-02308-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 01/22/2019] [Indexed: 12/28/2022]
Affiliation(s)
- Lorena P Suarez-Kelly
- Comprehensive Cancer Center, The Ohio State University, Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, N924 Doan Hall 410 W. 10th Ave, Columbus, OH, 43210-1228, USA
| | - Kala M Levine
- Comprehensive Cancer Center, The Ohio State University, Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, N924 Doan Hall 410 W. 10th Ave, Columbus, OH, 43210-1228, USA
| | - Thomas E Olencki
- Medical Oncology, Department of Internal Medicine, The Ohio State University, Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, Columbus, OH, USA
| | | | | | - Taylor R Brooks
- Division of Rheumatology and Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Volodymyr I Karpa
- Comprehensive Cancer Center, The Ohio State University, Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, N924 Doan Hall 410 W. 10th Ave, Columbus, OH, 43210-1228, USA
| | - Joseph Markowitz
- Comprehensive Cancer Center, The Ohio State University, Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, N924 Doan Hall 410 W. 10th Ave, Columbus, OH, 43210-1228, USA
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Anissa K Bingman
- Department of Biomedical Informatics, The Ohio State University College of Medicine, Columbus, OH, USA
- Hematology, Department of Internal Medicine, The Ohio State University, Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, Columbus, OH, USA
| | - Susan M Geyer
- Department of Biomedical Informatics, The Ohio State University College of Medicine, Columbus, OH, USA
- Hematology, Department of Internal Medicine, The Ohio State University, Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, Columbus, OH, USA
| | - Kari L Kendra
- Medical Oncology, Department of Internal Medicine, The Ohio State University, Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, Columbus, OH, USA
| | - William E Carson
- Comprehensive Cancer Center, The Ohio State University, Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, N924 Doan Hall 410 W. 10th Ave, Columbus, OH, 43210-1228, USA.
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
| |
Collapse
|
894
|
Rabbie R, Ferguson P, Molina‐Aguilar C, Adams DJ, Robles‐Espinoza CD. Melanoma subtypes: genomic profiles, prognostic molecular markers and therapeutic possibilities. J Pathol 2019; 247:539-551. [PMID: 30511391 PMCID: PMC6492003 DOI: 10.1002/path.5213] [Citation(s) in RCA: 147] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 11/27/2018] [Accepted: 11/30/2018] [Indexed: 12/24/2022]
Abstract
Melanoma is characterised by its ability to metastasise at early stages of tumour development. Current clinico-pathologic staging based on the American Joint Committee on Cancer criteria is used to guide surveillance and management in early-stage disease, but its ability to predict clinical outcome has limitations. Herein we review the genomics of melanoma subtypes including cutaneous, acral, uveal and mucosal, with a focus on the prognostic and predictive significance of key molecular aberrations. © 2018 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Roy Rabbie
- Experimental Cancer GeneticsThe Wellcome Sanger InstituteHinxtonUK
- Cambridge Cancer CentreCambridge University Hospitals NHS Foundation TrustCambridgeUK
| | - Peter Ferguson
- Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred HospitalSydneyAustralia
- Melanoma Institute Australia, The University of SydneySydneyAustralia
| | - Christian Molina‐Aguilar
- Laboratorio Internacional de Investigación sobre el Genoma HumanoUniversidad Nacional Autónoma de MéxicoSantiago de QuerétaroMexico
| | - David J Adams
- Experimental Cancer GeneticsThe Wellcome Sanger InstituteHinxtonUK
| | - Carla D Robles‐Espinoza
- Experimental Cancer GeneticsThe Wellcome Sanger InstituteHinxtonUK
- Laboratorio Internacional de Investigación sobre el Genoma HumanoUniversidad Nacional Autónoma de MéxicoSantiago de QuerétaroMexico
| |
Collapse
|
895
|
Coit DG, Thompson JA, Albertini MR, Barker C, Carson WE, Contreras C, Daniels GA, DiMaio D, Fields RC, Fleming MD, Freeman M, Galan A, Gastman B, Guild V, Johnson D, Joseph RW, Lange JR, Nath S, Olszanski AJ, Ott P, Gupta AP, Ross MI, Salama AK, Skitzki J, Sosman J, Swetter SM, Tanabe KK, Wuthrick E, McMillian NR, Engh AM. Cutaneous Melanoma, Version 2.2019, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw 2019; 17:367-402. [PMID: 30959471 DOI: 10.6004/jnccn.2019.0018] [Citation(s) in RCA: 290] [Impact Index Per Article: 48.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines) for Cutaneous melanoma have been significantly revised over the past few years in response to emerging data on immune checkpoint inhibitor therapies and BRAF-targeted therapy. This article summarizes the data and rationale supporting extensive changes to the recommendations for systemic therapy as adjuvant treatment of resected disease and as treatment of unresectable or distant metastatic disease.
Collapse
Affiliation(s)
| | - John A Thompson
- 2Fred Hutchinson Cancer Research Center/Seattle Cancer Care Alliance
| | | | | | - William E Carson
- 4The Ohio State University Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute
| | - Carlo Contreras
- 5University of Alabama at Birmingham Comprehensive Cancer Center
| | | | | | - Ryan C Fields
- 8Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine
| | - Martin D Fleming
- 9St. Jude Children's Research Hospital/The University of Tennessee Health Science Center
| | | | | | - Brian Gastman
- 12Case Comprehensive Cancer Center/University Hospitals Seidman Cancer Center and Cleveland Clinic Taussig Cancer Institute
| | | | | | | | - Julie R Lange
- 16The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins
| | | | | | - Patrick Ott
- 19Dana-Farber/Brigham and Women's Cancer Center
| | | | | | | | | | - Jeffrey Sosman
- 20Robert H. Lurie Comprehensive Cancer Center of Northwestern University
| | | | | | | | | | | |
Collapse
|
896
|
Schadendorf D, Hauschild A, Santinami M, Atkinson V, Mandalà M, Chiarion-Sileni V, Larkin J, Nyakas M, Dutriaux C, Haydon A, Robert C, Mortier L, Lesimple T, Plummer R, Schachter J, Dasgupta K, Manson S, Koruth R, Mookerjee B, Kefford R, Dummer R, Kirkwood JM, Long GV. Patient-reported outcomes in patients with resected, high-risk melanoma with BRAF V600E or BRAF V600K mutations treated with adjuvant dabrafenib plus trametinib (COMBI-AD): a randomised, placebo-controlled, phase 3 trial. Lancet Oncol 2019; 20:701-710. [PMID: 30928620 DOI: 10.1016/s1470-2045(18)30940-9] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 11/29/2018] [Accepted: 12/05/2018] [Indexed: 12/19/2022]
Abstract
BACKGROUND In the phase 3 COMBI-AD study, patients with resected, stage III melanoma with BRAFV600E or BRAFV600K mutations received adjuvant dabrafenib plus trametinib or placebo. The primary analysis showed that dabrafenib plus trametinib significantly improved relapse-free survival at 3 years. These results led to US Food and Drug Administration approval of dabrafenib plus trametinib as adjuvant treatment for patients with resected stage III melanoma with BRAFV600E or BRAFV600K mutations. Here, we report the patient-reported outcomes from COMBI-AD. METHODS COMBI-AD was a randomised, double-blind, placebo-controlled, phase 3 study done at 169 sites in 25 countries. Study participants were aged 18 years or older and had complete resection of stage IIIA (lymph node metastases >1 mm), IIIB, or IIIC cutaneous melanoma as per American Joint Committee on Cancer 7th edition criteria, with BRAFV600E or BRAFV600K mutations, and an Eastern Cooperative Oncology Group performance status of 0 or 1. Patients were randomly assigned (1:1) via an interactive voice response system, stratified by mutation type and disease stage, to receive oral dabrafenib (150 mg twice daily) plus oral trametinib (2 mg once daily) or matching placebos for 12 months. Patients, physicians, and the investigators who analysed the data were masked to treatment allocation. The primary endpoint was relapse-free survival, reported elsewhere. Health-related quality of life, reported here, was a prespecified exploratory endpoint, and was assessed with the European Quality of Life 5-Dimensions 3-Levels (EQ-5D-3L) questionnaire in the intention-to-treat population. We used a mixed-model repeated-measures analysis to assess differences in health-related quality of life between groups. This study is registered with ClinicalTrials.gov, number NCT01682083. The trial is ongoing, but is no longer recruiting participants. FINDINGS Between Jan 31, 2013, and Dec 11, 2014, 870 patients were enrolled and randomly assigned to receive dabrafenib plus trametinib (n=438) or matching placebos (n=432). Data were collected until the data cutoff for analyses of the primary endpoint (June 30, 2017). The median follow-up was 34 months (IQR 28-39) in the dabrafenib plus trametinib group and 33 months (20·5-39) in the placebo group. During the 12-month treatment phase, there were no significant or clinically meaningful changes from baseline between groups in EQ-5D-3L visual analogue scale (EQ-VAS) or utility scores. During treatment, there were no clinically meaningful differences in VAS scores or utility scores in the dabrafenib plus trametinib group between patients who did and did not experience the most common adverse events. During long-term follow-up (range 15-48 months), VAS and utility scores were similar between groups and did not differ from baseline scores. At recurrence, there were significant decreases in VAS scores in both the dabrafenib plus trametinib group (mean change -6·02, SD 20·57; p=0·0032) and the placebo group (-6·84, 20·86; p<0·0001); the mean change in utility score also differed significantly at recurrence for both groups (dabrafenib plus trametinib -0·0626, 0·1911, p<0·0001; placebo -0·0748, 0·2182, p<0·0001). INTERPRETATION These findings show that dabrafenib plus trametinib did not affect patient-reported outcome scores during or after adjuvant treatment, and suggest that preventing or delaying relapse with adjuvant therapy could be beneficial in this setting. FUNDING Novartis.
Collapse
Affiliation(s)
- Dirk Schadendorf
- Department of Dermatology, University Hospital Essen, Essen, Germany; German Cancer Consortium, Heidelberg, Germany.
| | | | | | - Victoria Atkinson
- Princess Alexandra Hospital, Gallipoli Medical Research Foundation, University of Queensland, QLD, Australia
| | - Mario Mandalà
- Papa Giovanni XXIII Cancer Center Hospital, Bergamo, Italy
| | | | | | - Marta Nyakas
- Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
| | - Caroline Dutriaux
- Centre Hospitalier Universitaire de Bordeaux, Hôpital Saint-André, Bordeaux, France
| | | | | | | | - Thierry Lesimple
- Medical Oncology Department, Centre Eugène Marquis, Rennes, France
| | - Ruth Plummer
- Northern Centre for Cancer Care, Freeman Hospital and Newcastle University, Newcastle upon Tyne, UK
| | - Jacob Schachter
- Ella Lemelbaum Institute for Immuno-Oncology and Melanoma, Sheba Medical Center, Tel Hashomer, Israel; Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | | | | | - Roy Koruth
- Novartis Pharmaceuticals, East Hanover, NJ, USA
| | | | - Richard Kefford
- Macquarie University, Sydney, NSW, Australia; Melanoma Institute Australia, Sydney, NSW, Australia; Westmead Hospital, Sydney, NSW, Australia; The University of Sydney, Sydney, NSW, Australia
| | - Reinhard Dummer
- University Hospital Zürich Skin Cancer Center, Zürich, Switzerland
| | - John M Kirkwood
- Melanoma Program, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Georgina V Long
- Melanoma Institute Australia, Sydney, NSW, Australia; The University of Sydney, Sydney, NSW, Australia; Royal North Shore Hospital, Sydney, NSW, Australia; Mater Hospital, Sydney, NSW, Australia
| |
Collapse
|
897
|
Angeles CV, Kang R, Shirai K, Wong SL. Meta-analysis of completion lymph node dissection in sentinel lymph node-positive melanoma. Br J Surg 2019; 106:672-681. [PMID: 30912591 DOI: 10.1002/bjs.11149] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 11/27/2018] [Accepted: 02/04/2019] [Indexed: 11/09/2022]
Abstract
BACKGROUND The role of completion lymph node dissection (CLND) in patients with sentinel lymph node (SLN)-positive melanoma continues to be debated. This systematic review and meta-analysis evaluated survival and recurrence rate in these patients who underwent CLND, compared with observation. METHODS A comprehensive MEDLINE and Embase database search was performed for cohort studies and RCTs published between January 2000 and June 2017 that assessed the outcomes of CLND compared with observation in patients with SLN-positive melanoma. The primary outcome was survival and the secondary outcome was recurrence rate. Studies were assessed for quality using the Cochrane risk-of-bias tool for RCTs and Newcastle-Ottawa Scale for cohort studies. Pooled relative risk or hazard ratio with 95 per cent confidence intervals were calculated for each outcome. The extent of heterogeneity between studies was assessed with the I2 test. The protocol was registered in PROSPERO (CRD42017070152). RESULTS Fifteen studies (13 cohort studies with 7868 patients and 2 RCTs with 2228 patients) were identified for qualitative synthesis. Thirteen studies remained for quantitative meta-analysis. Survival was similar in patients who underwent CLND and those who were observed (risk ratio (RR) for death 0·85, 95 per cent c.i. 0·71 to 1·02). The recurrence rate was also similar (RR 0·91, 0·79 to 1·05). CONCLUSION Patients with SLN-positive melanoma do not have a significant benefit in survival or recurrence rate if they undergo CLND rather than observation.
Collapse
Affiliation(s)
- C V Angeles
- Department of Surgery, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, USA
| | - R Kang
- Department of Surgery, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, USA
| | - K Shirai
- Department of Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, USA
| | - S L Wong
- Department of Surgery, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, USA
| |
Collapse
|
898
|
Nijhuis AAG, Dieng M, Khanna N, Lord SJ, Dalton J, Menzies AM, Turner RM, Allen J, Saw RPM, Nieweg OE, Thompson JF, Morton RL. False-Positive Results and Incidental Findings with Annual CT or PET/CT Surveillance in Asymptomatic Patients with Resected Stage III Melanoma. Ann Surg Oncol 2019; 26:1860-1868. [DOI: 10.1245/s10434-019-07311-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Indexed: 11/18/2022]
|
899
|
Savoia P, Fava P, Casoni F, Cremona O. Targeting the ERK Signaling Pathway in Melanoma. Int J Mol Sci 2019; 20:ijms20061483. [PMID: 30934534 PMCID: PMC6472057 DOI: 10.3390/ijms20061483] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 03/17/2019] [Accepted: 03/19/2019] [Indexed: 12/24/2022] Open
Abstract
The discovery of the role of the RAS/RAF/MEK/ERK pathway in melanomagenesis and its progression have opened a new era in the treatment of this tumor. Vemurafenib was the first specific kinase inhibitor approved for therapy of advanced melanomas harboring BRAF-activating mutations, followed by dabrafenib and encorafenib. However, despite the excellent results of first-generation kinase inhibitors in terms of response rate, the average duration of the response was short, due to the onset of genetic and epigenetic resistance mechanisms. The combination therapy with MEK inhibitors is an excellent strategy to circumvent drug resistance, with the additional advantage of reducing side effects due to the paradoxical reactivation of the MAPK pathway. The recent development of RAS and extracellular signal-related kinases (ERK) inhibitors promises to add new players for the ultimate suppression of this signaling pathway and the control of pathway-related drug resistance. In this review, we analyze the pharmacological, preclinical, and clinical trial data of the various MAPK pathway inhibitors, with a keen interest for their clinical applicability in the management of advanced melanoma.
Collapse
Affiliation(s)
- Paola Savoia
- Department of Health Science, University of Eastern Piedmont, via Solaroli 17, 28100 Novara, Italy.
| | - Paolo Fava
- Section of Dermatology, Department of Medical Science, University of Turin, 10124 Turin, Italy.
| | - Filippo Casoni
- San Raffaele Scientific Institute, Division of Neuroscience, via Olgettina 58, 20132 Milano, Italy.
- Università Vita Salute San Raffaele, via Olgettina 58, 20132 Milano, Italy.
| | - Ottavio Cremona
- San Raffaele Scientific Institute, Division of Neuroscience, via Olgettina 58, 20132 Milano, Italy.
- Università Vita Salute San Raffaele, via Olgettina 58, 20132 Milano, Italy.
| |
Collapse
|
900
|
Verver D, van der Veldt A, van Akkooi A, Verhoef C, Grünhagen DJ, Louwman WJ. Treatment of melanoma of unknown primary in the era of immunotherapy and targeted therapy: A Dutch population-based study. Int J Cancer 2019; 146:26-34. [PMID: 30801710 PMCID: PMC6900034 DOI: 10.1002/ijc.32229] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 02/23/2019] [Accepted: 01/30/2019] [Indexed: 12/18/2022]
Abstract
Melanoma of unknown primary (MUP) may have a different biology to melanoma of known primary, but clinical trials of novel therapies (e.g., immune checkpoint or BRAF/MEK inhibitors) have not reported the outcomes in this population. We therefore evaluated the overall survival (OS) among patients with MUP in the era of novel therapy. Data for stage III or IV MUP were extracted from a nationwide database for the period 2003–2016, with classification based on the eighth edition of the American Joint Committee on Cancer criteria. The population was divided into pre‐ (2003–2010) and post‐ (2011–2016) novel therapy eras. Also, OS in the post‐novel era was compared between patients with stage IV MUP by whether they received novel therapy. In total, 2028 of 65,110 patients (3.1%) were diagnosed with MUP. Metastatic sites were known in 1919 of 2028 patients, and most had stage IV disease (53.8%). For patients with stage III MUP, the 5‐year OS rates were 48.5% and 50.2% in the pre‐ and post‐novel eras, respectively (p = 0.948). For those with stage IV MUP, the median OS durations were unchanged in the pre‐novel era and post‐novel era when novel therapy was not used (both 4 months); however, OS improved to 11 months when novel therapy was used in the post‐novel era (p < 0.001). In conclusion, more than half of the patients with MUP are diagnosed with stage IV and the introduction of novel therapy appears to have significantly improved the OS of these patients. What's new? Melanoma of unknown primary (MUP) site may have a different biology to melanoma of known primary, but clinical trials of novel therapies (e.g., immune checkpoint or BRAF/MEK inhibitors) have not reported the outcomes in this population. Knowledge about outcomes could however aid clinical management of patients with MUP. In this nationwide study from 2003 to 2016, the authors show that the introduction of novel therapy has significantly improved the overall survival for patients with stage IV melanoma of unknown primary, who represented more than half of the patients diagnosed with MUP in the Netherlands.
Collapse
Affiliation(s)
- D Verver
- Department of Surgical Oncology, Erasmus MC Cancer Institute, EA Rotterdam, The Netherlands
| | - Aam van der Veldt
- Department of Medical Oncology and Radiology & Nuclear Medicine, Erasmus MC Cancer Institute, EA Rotterdam, The Netherlands
| | - Acj van Akkooi
- Department of Surgical Oncology, Netherlands Cancer Institute-Antoni van Leeuwenhoek, CX Amsterdam, The Netherlands
| | - C Verhoef
- Department of Surgical Oncology, Erasmus MC Cancer Institute, EA Rotterdam, The Netherlands
| | - D J Grünhagen
- Department of Surgical Oncology, Erasmus MC Cancer Institute, EA Rotterdam, The Netherlands
| | - W J Louwman
- Department of Research, Netherlands Comprehensive Cancer Organization (IKNL), DB Utrecht, The Netherlands
| |
Collapse
|