151
|
Abstract
PURPOSE OF REVIEW Neoadjuvant therapy in melanoma is an area of active investigation with numerous completed and ongoing trials studying a variety of therapeutic interventions utilizing diverse designs. Here, we review completed and ongoing neoadjuvant trials in melanoma, discuss endpoint assessment, and highlight biomarker development in this context. RECENT FINDINGS High-risk resectable melanoma with clinically detectable lymph node (LN) with or without in-transit and/or satellite metastases represent ~ 20% of melanoma patients and have a high risk of relapse despite definitive surgery. Adjuvant therapy with anti-PD-1 immunotherapy or BRAF/MEK-targeted therapy has improved relapse-free survival (RFS) and overall survival (OS) in large phase III trials and is approved for this indication. However, despite surgery and adjuvant therapy, many patients relapse and/or experience treatment-related toxicity, underscoring the need to identify and understand mechanisms of response and resistance. In melanoma, neoadjuvant therapy is an active area of research with numerous completed and ongoing trials utilizing FDA-approved and novel agents with intriguing results. Neoadjuvant therapy for regionally metastatic disease is an established standard in multiple cancers, where it has been shown to improve operability, facilitate biomarker development, and even is a registrational endpoint for drug development in breast cancer. Recently, a spate of neoadjuvant studies in melanoma has looked at a swathe of agents with promising clinical and biomarker results. Coordinated efforts are underway to translate these findings to earlier stage disease while prioritizing the evaluation of new strategies in unresectable disease.
Collapse
|
152
|
Jang S, Poretta T, Bhagnani T, Harshaw Q, Burke M, Rao S. Real-World Recurrence Rates and Economic Burden in Patients with Resected Early-Stage Melanoma. Dermatol Ther (Heidelb) 2020; 10:985-999. [PMID: 32548707 PMCID: PMC7477064 DOI: 10.1007/s13555-020-00404-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Indexed: 01/27/2023] Open
Abstract
Introduction Real-world data on recurrence and economic burden in patients with resected early-stage melanoma are limited. The objective of this study was to assess real-world recurrence rates, risk factors for recurrence, and costs of recurrence in patients with resected stage IIB, IIC, or IIIA melanoma in the USA. Methods This retrospective analysis included patients with resected stage IIB, IIC, or IIIA melanoma (American Joint Committee on Cancer staging manual, seventh edition) in the Surveillance, Epidemiology, and End Results (SEER) program–Medicare database of the National Cancer Institute. Recurrence rates and healthcare costs (2018 USD) after recurrence were assessed. Results Two-year recurrence rates for stages IIB, IIC, and IIIA melanoma were 29, 44, and 46%, respectively. In patients with stage IIB or IIC disease, the odds of recurrence were significantly higher in those aged > 75 years [odds ratio (OR) 1.853, 95% confidence interval (CI) 1.416, 2.425], with ulceration (OR 1.771; 95% CI 1.293, 2.425), or with a higher Charlson Comorbidity Index (OR 1.244; 95% CI 1.129, 1.372); however, the odds of recurrence were significantly lower in those with T3 staging (OR 0.522; 95% CI 0.393, 0.695). In those with stage IIIA melanoma, superficial spreading was associated with significantly lower odds of recurrence (OR 0.178; 95% CI 0.053, 0.601). Following recurrence, mean healthcare costs at 1 year were $31,870 for patients with stage IIB or IIC melanoma and $29,224 for those with stage IIIA melanoma. Conclusion The SEER data show that a substantial proportion of adults with early-stage melanoma experience a recurrence within 2 years following resection, resulting in a significant economic burden to the US healthcare system. Dermatologists can distinguish patients with resected early-stage melanoma who are at a high risk for recurrence and consider referrals to medical oncologists for approved adjuvant therapy or enrollment in clinical trials after surgical resection to reduce the recurrence of melanoma. Melanoma is the sixth most common type of cancer in the USA. In the past several years, the US Food and Drug Administration has approved several novel therapies for patients with high-risk melanoma following surgery. However, these therapies are not approved for the treatment of patients with earlier-stage or intermediate-risk melanoma. In these patients, treatment choices include enrollment in clinical trials or observation. We have assessed recurrence rates, risk factors for recurrence, and costs of recurrence in patients with early-stage melanoma. This analysis included patients with resected early-stage melanoma in the US Surveillance, Epidemiology, and End Results (SEER) program–Medicare database. The results show that a substantial proportion of adults with early-stage melanoma experienced a recurrence within 2 years after surgical removal of their tumor, resulting in a significant economic burden to the healthcare system. Dermatologists can use information in the published literature to distinguish patients with resected early-stage melanoma who are at a high risk for recurrence and consider referrals to medical oncologists for approved therapy or enrollment in clinical trials after surgery to reduce recurrence and substantial economic consequences.
Collapse
Affiliation(s)
- Sekwon Jang
- Melanoma and Cutaneous Oncology Therapeutics, Inova Schar Cancer Institute, Fairfax, VA, USA.
| | - Tayla Poretta
- Health Economics and Outcomes Research, Bristol Myers Squibb, Princeton, NJ, USA
| | - Tarun Bhagnani
- Health Economics and Outcomes Research, EPI-Q Inc., Oakbrook, IL, USA
| | - Qing Harshaw
- Health Economics and Outcomes Research, EPI-Q Inc., Oakbrook, IL, USA
| | - Matthew Burke
- Health Economics and Outcomes Research, Bristol Myers Squibb, Princeton, NJ, USA
| | - Sumati Rao
- Health Economics and Outcomes Research, Bristol Myers Squibb, Princeton, NJ, USA
| |
Collapse
|
153
|
Franke V, van Akkooi ACJ. The extent of surgery for stage III melanoma: how much is appropriate? Lancet Oncol 2020; 20:e167-e174. [PMID: 30842060 DOI: 10.1016/s1470-2045(19)30099-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 01/15/2019] [Accepted: 01/16/2019] [Indexed: 12/19/2022]
Abstract
Since the first documented lymph node dissection in 1892, many trials have investigated the potential effect of this surgical procedure on survival in patients with melanoma. Two randomised controlled trials were unable to demonstrate improved survival with completion lymph node dissection versus nodal observation in patients with sentinel node-positive disease, although patients with larger sentinel node metastases (>1 mm) might benefit more from observation than from dissection, and could potentially be considered for adjuvant systemic therapy instead of complete dissection. Adjuvant immunotherapy with high-dose ipilimumab has led to improvements in overall survival, whereas therapy with nivolumab and pembrolizumab has improved relapse-free survival with greater safety. Furthermore, adjuvant-targeted therapy with dabrafenib and trametinib has improved survival outcomes in BRAFV600E and BRAFV600K-mutated melanomas. Three neoadjuvant trials have all shown high response rates, including complete responses, after short-term combination therapy with ipilimumab and nivolumab with no recurrences so far, although follow-up is still short. Despite the absence of a survival benefit with completion lymph node dissection in patients with sentinel node-positive or negative disease, the use of sentinel node staging will increase because of the introduction of effective adjuvant therapies. However, routine completion lymph node dissection for sentinel node-positive disease should be reconsidered. Accordingly, existing clinical guidelines are currently being revised. For palpable (macroscopic) nodal disease, the type and extent of surgery could be reduced if the index node can accurately predict the response and if studies show that lymph node dissection can be safely foregone in patients with a complete response. Overall, the appropriate type and extent of surgery for stage III melanoma is changing and becoming more personalised.
Collapse
Affiliation(s)
- Viola Franke
- Department of Surgical Oncology, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, Netherlands
| | - Alexander C J van Akkooi
- Department of Surgical Oncology, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, Netherlands.
| |
Collapse
|
154
|
Abstract
In resected high-risk melanoma (stage IIB/C-III) the risk of locoregional and/or distant recurrence is substantial and so far adjuvant therapies have been fairly unsuccessful. Interferon showed slight improvements in recurrence-free survival (RFS) but failed to convincingly improve overall survival (OS). In these patients, adjuvant therapy with treatments that show promising results in stage IV disease is arising. Studies using immune checkpoint blockade with anti-CTLA-4 and anti-PD-1 agents reveal convincing RFS benefits. OS rates, however, are not mature yet in most studies. Only ipilimumab has shown an OS benefit but at a high cost of toxicity. Also in studies with adjuvant targeted therapy using BRAF and MEK inhibitors, ensuring results are reported regarding RFS. As possible toxicity cannot be ignored, it is crucial to identify patients who would benefit most from these adjuvant therapies. In patients with clinically detectable lymph node metastases, studies using neoadjuvant schedules of immunotherapy and targeted therapy have been performed. In phase I and II studies the most optimal schedule of combination immunotherapy was identified and further research on this front will follow in the coming years. Concluding, after decades of scarce options for patients with high-risk melanoma, recent developments in adjuvant therapy have changed the standard of care for these patients.
Collapse
|
155
|
Hillen LM, Geybels MS, Spassova I, Becker JC, Gambichler T, Garmyn M, Zur Hausen A, van den Oord J, Winnepenninckx V. A digital mRNA expression signature to classify challenging Spitzoid melanocytic neoplasms. FEBS Open Bio 2020; 10:1326-1341. [PMID: 32431053 PMCID: PMC7327909 DOI: 10.1002/2211-5463.12897] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 04/21/2020] [Accepted: 05/15/2020] [Indexed: 12/19/2022] Open
Abstract
Spitzoid neoplasms are a challenging group of cutaneous melanocytic proliferations. They are characterized by epithelioid and/or spindle-shaped melanocytes and classified as benign Spitz nevi (SN), atypical Spitz tumors (AST), or malignant Spitz tumors (MST). The intermediate AST category represents a diagnostically challenging group since on purely histopathological grounds, their benign or malignant character remains unpredictable. This results in uncertainties in patient treatment and prognosis. The molecular properties of Spitzoid lesions, especially their transcriptomic landscape, remain poorly understood, and genomic alterations in melanoma-associated oncogenes are typically absent. The aim of this study was to characterize their transcriptome with digital mRNA expression profiling. Formalin-fixed paraffin-embedded samples (including 27 SN, 10 AST, and 14 MST) were analyzed using the NanoString nCounter PanCancer Pathways Panel. The number of significantly differentially expressed genes in SN vs. MST, SN vs. AST, and AST vs. MST was 68, 167, and 18, respectively. Gene set enrichment analysis revealed upregulation of pathways related to epithelial-mesenchymal transition and immunomodulatory-, angiogenesis-, hormonal-, and myogenesis-associated processes in AST and MST. A molecular signature of SN vs. MST was discovered based on the top-ranked most informative genes: NRAS, NF1, BMP2, EIF2B4, IFNA17, and FZD9. The AST samples showed intermediate levels of the identified signature. This implies that the gene signature can potentially be used to distinguish high-grade from low-grade AST with a larger study cohort in the future. This combined histopathological and transcriptomic methodology is promising for prospective diagnostics of Spitzoid neoplasms and patient management in dermatological oncology.
Collapse
Affiliation(s)
- Lisa M Hillen
- Department of Pathology, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center (MUMC+), Maastricht, the Netherlands
| | - Milan S Geybels
- Department of Epidemiology, GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht, the Netherlands
| | - Ivelina Spassova
- Department for Translational Skin Cancer Research (TSCR), German Cancer Consortium (DKTK), University Hospital Essen, Essen, Germany
| | - Jürgen C Becker
- Department for Translational Skin Cancer Research (TSCR), German Cancer Consortium (DKTK), University Hospital Essen, Essen, Germany.,Deutsches Krebsforschungsinstitut (DKFZ), Heidelberg, Germany
| | - Thilo Gambichler
- Department of Dermatology, Ruhr-University Bochum, Bochum, Germany
| | - Marjan Garmyn
- Laboratory of Dermatology, Department of Oncology and Department of Dermatology, University Hospitals Leuven, University of Leuven KUL, Leuven, Belgium
| | - Axel Zur Hausen
- Department of Pathology, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center (MUMC+), Maastricht, the Netherlands
| | - Joost van den Oord
- Department of Pathology, University Hospitals of Leuven, University of Leuven KUL, Leuven, Belgium.,Laboratory Translational Cell and Tissue Research, University of Leuven, KU, Leuven, Belgium
| | - Véronique Winnepenninckx
- Department of Pathology, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center (MUMC+), Maastricht, the Netherlands
| |
Collapse
|
156
|
Eggermont AMM, Rutkowski P, Dutriaux C, Hofman-Wellenhof R, Dziewulski P, Marples M, Grange F, Lok C, Pennachioli E, Robert C, van Akkooi ACJ, Bastholt L, Minisini A, Marshall E, Salès F, Grob JJ, Bechter O, Schadendorf D, Marreaud S, Kicinski M, Suciu S, Testori AAE. Adjuvant therapy with pegylated interferon-alfa2b vs observation in stage II B/C patients with ulcerated primary: Results of the European Organisation for Research and Treatment of Cancer 18081 randomised trial. Eur J Cancer 2020; 133:94-103. [PMID: 32470710 DOI: 10.1016/j.ejca.2020.04.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 04/10/2020] [Indexed: 11/28/2022]
Abstract
BACKGROUND Subgroup analyses of two large EORTC adjuvant interferon-alpha2b (IFNα-2b) vs observation randomised trials demonstrated that a treatment benefit was observed only in patients with an ulcerated melanoma without palpable nodes (hazard ratio [HR] for recurrence-free survival [RFS] was 0.69). This was confirmed by a meta-analysis of 15 adjuvant IFN trials (HR: 0.79). PATIENTS AND METHODS In the EORTC 18081 trial, sentinel node-negative stage II patients with an ulcerated primary melanoma were 1:1 randomised between pegylated (PEG)-IFNα-2b at 3 μg/kg/week subcutaneously and observation, for 2 years, or until disease recurrence or unacceptable toxicity in spite of dose adjustments to maintain an Eastern Cooperative Oncology Group performance status of 0 or 1. Main end-point was RFS. Secondary end-points included distant metastasis-free survival (DMFS), overall survival, and safety (EudraCT Number: 2009-010273-20). RESULTS Between February 2013 and January 2017, only 112 patients were randomised, 56 in each arm. The trial was stopped early for lack of recruitment. At a 3.4-year median follow-up, the estimated HR for the PEG-IFNα-2b group compared with the observation group regarding RFS was 0.66 (95% confidence interval [CI]: 0.32-1.37), and the 3-year RFS rate was 80.0% (95% CI: 65.7-88.8%) and 72.9% (95% CI: 58.3-83.0%), respectively. DMFS was prolonged: HR: 0.39 (95% CI: 0.15-0.97), and the 3-year DMFS rate was 90.6% (95% CI: 78.9-96.0%) vs 76.4% (95% CI: 62.1-85.9%). One patient in the PEG-IFNα-2b group died compared with 4 in the observation group. Fifty-four patients started PEG-IFNα-2b treatment, 16 (29%) completed 2 years of treatment, 2 (4%) stopped due to recurrence, 23 (43%) due to toxicity and 14 (25%) due to other reasons. CONCLUSIONS The EORTC 18081 PEG-IFNα-2b randomised trial, observed a similar HR (0.69) for RFS as the previous EORTC trials (0.69). In countries without access to new drugs, adjuvant (PEG)-IFNα-2b treatment is an option for patients with ulcerated melanomas without palpable nodes.
Collapse
Affiliation(s)
| | - Piotr Rutkowski
- Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland.
| | - Caroline Dutriaux
- CHU de Bordeaux, Groupe Hospitalier Saint-André, Hopital Saint-André, Bordeaux, France.
| | | | - Peter Dziewulski
- Mid Essex Hospitals, Broomfield Hospital, Broomfield, United Kingdom.
| | - Maria Marples
- Leeds Teaching Hospitals NHS Trust, St. James's University Hospital, Leeds, United Kingdom.
| | | | | | | | - Caroline Robert
- Gustave Roussy, Villejuif & Paris-Saclay University, Saint-Aubin, France.
| | | | | | | | - Ernest Marshall
- St Helens & Knowsley NHS Trust, Whiston Hospital, Prescot, United Kingdom.
| | - François Salès
- Institut Jules Bordet-Hopital Universitaire ULB, Brussels, Belgium.
| | - Jean-Jacques Grob
- Assistance Publique, Hopitaux de Marseille, Hôpital de La Timone (APHM), Marseille, France.
| | - Oliver Bechter
- Department of General Medical Oncology, University Hospitals Leuven, Campus Gasthuisberg, Leuven, Belgium.
| | - Dirk Schadendorf
- University Hospital Essen, Essen & German Cancer Consortium, Heidelberg, Germany.
| | | | | | | | | |
Collapse
|
157
|
Predicting circulating biomarker response and its impact on the survival of advanced melanoma patients treated with adjuvant therapy. Sci Rep 2020; 10:7478. [PMID: 32366871 PMCID: PMC7198615 DOI: 10.1038/s41598-020-63441-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 03/30/2020] [Indexed: 11/21/2022] Open
Abstract
Advanced melanoma remains a disease with poor prognosis. Several serologic markers have been investigated to help monitoring and prognostication, but to date only lactate dehydrogenase (LDH) has been validated as a standard prognostic factor biomarker for this disease by the American Joint Committee on Cancer. In this work, we built a semi-mechanistic model to explore the relationship between the time course of several circulating biomarkers and overall or progression free survival in advanced melanoma patients treated with adjuvant high-dose interferon-\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$${\boldsymbol{\alpha }}{\bf{2}}{\bf{b}}$$\end{document}α2b. Additionally, due to the adverse interferon tolerability, a semi-mechanistic model describing the side effects of the treatment in the absolute neutrophil counts is proposed in order to simultaneously analyze the benefits and toxic effects of this treatment. The results of our analysis suggest that the relative change from baseline of LDH was the most significant predictor of the overall survival of the patients. Unfortunately, there was no significant difference in the proportion of patients with elevated serum biomarkers between the patients who recurred and those who remained free of disease. Still, we believe that the modelling framework presented in this work of circulating biomarkers and adverse effects could constitute an additional strategy for disease monitoring in advance melanoma patients.
Collapse
|
158
|
Burke K, Jones MC, Noufaily A. A flexible parametric modelling framework for survival analysis. J R Stat Soc Ser C Appl Stat 2020. [DOI: 10.1111/rssc.12398] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
159
|
Krattinger R, Ramelyte E, Dornbierer J, Dummer R. Is single versus combination therapy problematic in the treatment of cutaneous melanoma? Expert Rev Clin Pharmacol 2020; 14:9-23. [PMID: 31364890 DOI: 10.1080/17512433.2019.1650641] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Introduction: The development of immunotherapies and targeted therapies has changed the treatment approach in resectable, nonresectable, and metastatic melanoma. Because of their different pharmacological profiles, immunotherapies and/or targeted therapies have been studied in various combinations. Areas covered: We reviewed PubMed for most important clinical trials investigating efficacy and tolerability of combinatorial and single-agent approaches for the treatment of melanoma that were published up to June 2019. We discuss the most promising therapy approaches and highlight challenges of melanoma treatment. Expert opinion: Combinatorial approaches seem to be very promising in the treatment of resectable and advanced melanoma. Currently, dual immune checkpoint inhibition (ICI) with nivolumab and ipilimumab offers the best first-line treatment option for patients with BRAF-wt and -mutated, advanced melanoma. It is therapy of choice in younger patients with good ECOG performance status and poor prognostic features, whereas ICI monotherapy should be preferred in elderly patients with advanced melanoma. Benefit-risk ratio, patient's QoL and expectations, as well as treatment costs have to be considered in the choice of treatment. However, to elucidate mechanisms of resistance, biomarkers of response and to better define personalized strategies in the treatment of cutaneous melanoma, larger clinical trials comparing combined versus sequential therapies are necessary.
Collapse
Affiliation(s)
- Regina Krattinger
- Department of Dermatology, University Hospital Zurich , Zurich, Switzerland
| | - Egle Ramelyte
- Department of Dermatology, University Hospital Zurich , Zurich, Switzerland
| | - Joëlle Dornbierer
- Department of Dermatology, University Hospital Zurich , Zurich, Switzerland
| | - Reinhard Dummer
- Department of Dermatology, University Hospital Zurich , Zurich, Switzerland
| |
Collapse
|
160
|
Lamberti MJ, Nigro A, Mentucci FM, Rumie Vittar NB, Casolaro V, Dal Col J. Dendritic Cells and Immunogenic Cancer Cell Death: A Combination for Improving Antitumor Immunity. Pharmaceutics 2020; 12:pharmaceutics12030256. [PMID: 32178288 PMCID: PMC7151083 DOI: 10.3390/pharmaceutics12030256] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 03/05/2020] [Accepted: 03/10/2020] [Indexed: 02/07/2023] Open
Abstract
The safety and feasibility of dendritic cell (DC)-based immunotherapies in cancer management have been well documented after more than twenty-five years of experimentation, and, by now, undeniably accepted. On the other hand, it is equally evident that DC-based vaccination as monotherapy did not achieve the clinical benefits that were predicted in a number of promising preclinical studies. The current availability of several immune modulatory and targeting approaches opens the way to many potential therapeutic combinations. In particular, the evidence that the immune-related effects that are elicited by immunogenic cell death (ICD)-inducing therapies are strictly associated with DC engagement and activation strongly support the combination of ICD-inducing and DC-based immunotherapies. In this review, we examine the data in recent studies employing tumor cells, killed through ICD induction, in the formulation of anticancer DC-based vaccines. In addition, we discuss the opportunity to combine pharmacologic or physical therapeutic approaches that can promote ICD in vivo with in situ DC vaccination.
Collapse
Affiliation(s)
- María Julia Lamberti
- Departamento de Biología Molecular, Universidad Nacional de Río Cuarto, Río Cuarto 5800, Córdoba, Argentina; (M.J.L.); (F.M.M.)
- INBIAS, CONICET-UNRC, Río Cuarto 5800, Córdoba, Argentina
| | - Annunziata Nigro
- Department of Medicine, Surgery and Dentistry ‘Scuola Medica Salernitana’, University of Salerno, 84081 Baronissi, Salerno, Italy; (A.N.); (V.C.)
| | - Fátima María Mentucci
- Departamento de Biología Molecular, Universidad Nacional de Río Cuarto, Río Cuarto 5800, Córdoba, Argentina; (M.J.L.); (F.M.M.)
- INBIAS, CONICET-UNRC, Río Cuarto 5800, Córdoba, Argentina
| | - Natalia Belén Rumie Vittar
- Departamento de Biología Molecular, Universidad Nacional de Río Cuarto, Río Cuarto 5800, Córdoba, Argentina; (M.J.L.); (F.M.M.)
- INBIAS, CONICET-UNRC, Río Cuarto 5800, Córdoba, Argentina
- Correspondence: (N.B.R.V.); (J.D.C.); Tel.: +39-089-965-210 (J.D.C.)
| | - Vincenzo Casolaro
- Department of Medicine, Surgery and Dentistry ‘Scuola Medica Salernitana’, University of Salerno, 84081 Baronissi, Salerno, Italy; (A.N.); (V.C.)
| | - Jessica Dal Col
- Department of Medicine, Surgery and Dentistry ‘Scuola Medica Salernitana’, University of Salerno, 84081 Baronissi, Salerno, Italy; (A.N.); (V.C.)
- Correspondence: (N.B.R.V.); (J.D.C.); Tel.: +39-089-965-210 (J.D.C.)
| |
Collapse
|
161
|
Bashraheel SS, Domling A, Goda SK. Update on targeted cancer therapies, single or in combination, and their fine tuning for precision medicine. Biomed Pharmacother 2020; 125:110009. [PMID: 32106381 DOI: 10.1016/j.biopha.2020.110009] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Revised: 02/04/2020] [Accepted: 02/12/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Until recently, patients who have the same type and stage of cancer all receive the same treatment. It has been established, however, that individuals with the same disease respond differently to the same therapy. Further, each tumor undergoes genetic changes that cause cancer to grow and metastasize. The changes that occur in one person's cancer may not occur in others with the same cancer type. These differences also lead to different responses to treatment. Precision medicine, also known as personalized medicine, is a strategy that allows the selection of a treatment based on the patient's genetic makeup. In the case of cancer, the treatment is tailored to take into account the genetic changes that may occur in an individual's tumor. Precision medicine, therefore, could be defined in terms of the targets involved in targeted therapy. METHODS A literature search in electronic data bases using keywords "cancer targeted therapy, personalized medicine and cancer combination therapies" was conducted to include papers from 2010 to June 2019. RESULTS Recent developments in strategies of targeted cancer therapy were reported. Specifically, on the two types of targeted therapy; first, immune-based therapy such as the use of immune checkpoint inhibitors (ICIs), immune cytokines, tumor-targeted superantigens (TTS) and ligand targeted therapeutics (LTTs). The second strategy deals with enzyme/small molecules-based therapies, such as the use of a proteolysis targeting chimera (PROTAC), antibody-drug conjugates (ADC) and antibody-directed enzyme prodrug therapy (ADEPT). The precise targeting of the drug to the gene or protein under attack was also investigated, in other words, how precision medicine can be used to tailor treatments. CONCLUSION The conventional therapeutic paradigm for cancer and other diseases has focused on a single type of intervention for all patients. However, a large literature in oncology supports the therapeutic benefits of a precision medicine approach to therapy as well as combination therapies.
Collapse
Affiliation(s)
- Sara S Bashraheel
- Protein Engineering Unit, Life and Science Research Department, Anti-Doping Lab-Qatar (ADLQ), Doha, Qatar; Drug Design Group, Department of Pharmacy, University of Groningen, Groningen, Netherlands
| | - Alexander Domling
- Drug Design Group, Department of Pharmacy, University of Groningen, Groningen, Netherlands
| | - Sayed K Goda
- Cairo University, Faculty of Science, Chemistry Department, Giza, Egypt.
| |
Collapse
|
162
|
Normalization Cancer Immunotherapy for Melanoma. J Invest Dermatol 2020; 140:1134-1142. [PMID: 32092349 DOI: 10.1016/j.jid.2020.02.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 12/26/2019] [Accepted: 02/05/2020] [Indexed: 12/28/2022]
Abstract
Today, we are witnessing a revolution in the treatment of cancer using immunotherapy. In the past decade, work from many laboratories and clinicians has unequivocally demonstrated that the immune system can eradicate established cancers and enhance patient survival. However, immunotherapies have distinct tumor response-to-toxicity profiles owing to distinct mechanisms of action. We have previously termed immunotherapies that activate a general systemic immune response as enhancement cancer immunotherapy and those that target a specific dysfunctional immune response, especially within the tumor microenvironment, as normalization cancer immunotherapy. In this perspective, we provide a framework for normalization cancer immunotherapy in the context of melanoma.
Collapse
|
163
|
Stalin J, Traboulsi W, Vivancos-Stalin L, Nollet M, Joshkon A, Bachelier R, Guillet B, Lacroix R, Foucault-Bertaud A, Leroyer AS, Dignat-George F, Bardin N, Blot-Chabaud M. Therapeutic targeting of soluble CD146/MCAM with the M2J-1 monoclonal antibody prevents metastasis development and procoagulant activity in CD146-positive invasive tumors. Int J Cancer 2020; 147:1666-1679. [PMID: 32022257 DOI: 10.1002/ijc.32909] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 12/20/2019] [Accepted: 01/22/2020] [Indexed: 12/17/2022]
Abstract
Initially discovered in human melanoma, CD146/MCAM is expressed on many tumors and is correlated with cancer progression and metastasis. However, targeting CD146 remains challenging since it is also expressed on other cell types, as vessel cells, where it displays important physiological functions. We previously demonstrated that CD146 is shed as a soluble form (sCD146) that vectorizes the effects of membrane CD146 on tumor angiogenesis, growth and survival. We thus generated a novel monoclonal antibody, the M2J-1 mAb, which specifically targets sCD146, but not membrane CD146, and counteracts these effects. In our study, we analyzed the effects of sCD146 on the dissemination and the associated procoagulant phenotype in two highly invasive human CD146-positive cancer cell lines (ovarian and melanoma). Results show that sCD146 induced epithelial to mesenchymal transition, favored the generation of cancer stem cells and increased the membrane expression of tissue factor. Treatment of cancer cells with sCD146 in two experimental models (subcutaneous xenografting and intracardiac injection of cancer cells in nude mice) led to increased tumor dissemination and procoagulant activity. The M2J-1 mAb drastically reduced metastasis but also procoagulant activity, in particular by decreasing the number of circulating tumor microparticles, and blocked the relevant signaling pathways as demonstrated by RNA expression profiling experiments. Thus, our findings demonstrate that sCD146 mediates important pro-metastatic and procoagulant effects in two CD146-positive tumors. Targeting sCD146 with the newly generated M2J-1 mAb could constitute an innovative strategy for preventing dissemination and thromboembolism in many CD146-positive tumors.
Collapse
Affiliation(s)
- Jimmy Stalin
- INSERM, INRAE, C2VN, UFR Pharmacie, Aix-Marseille University, Marseille, France
| | - Wael Traboulsi
- INSERM, INRAE, C2VN, UFR Pharmacie, Aix-Marseille University, Marseille, France
| | | | - Marie Nollet
- INSERM, INRAE, C2VN, UFR Pharmacie, Aix-Marseille University, Marseille, France
| | - Ahmad Joshkon
- INSERM, INRAE, C2VN, UFR Pharmacie, Aix-Marseille University, Marseille, France
| | - Richard Bachelier
- INSERM, INRAE, C2VN, UFR Pharmacie, Aix-Marseille University, Marseille, France
| | - Benjamin Guillet
- INSERM, INRAE, C2VN, UFR Pharmacie, Aix-Marseille University, Marseille, France.,CERIMED (European Center of Research in Medical Imaging), Aix-Marseille University, Marseille, France
| | - Romaric Lacroix
- INSERM, INRAE, C2VN, UFR Pharmacie, Aix-Marseille University, Marseille, France.,AP-HM, La Conception Hospital, Marseille, France
| | | | - Aurélie S Leroyer
- INSERM, INRAE, C2VN, UFR Pharmacie, Aix-Marseille University, Marseille, France
| | - Françoise Dignat-George
- INSERM, INRAE, C2VN, UFR Pharmacie, Aix-Marseille University, Marseille, France.,AP-HM, La Conception Hospital, Marseille, France
| | - Nathalie Bardin
- INSERM, INRAE, C2VN, UFR Pharmacie, Aix-Marseille University, Marseille, France.,AP-HM, La Conception Hospital, Marseille, France
| | - Marcel Blot-Chabaud
- INSERM, INRAE, C2VN, UFR Pharmacie, Aix-Marseille University, Marseille, France
| |
Collapse
|
164
|
Tarhini AA, Lee SJ, Hodi FS, Rao UNM, Cohen GI, Hamid O, Hutchins LF, Sosman JA, Kluger HM, Eroglu Z, Koon HB, Lawrence DP, Kendra KL, Minor DR, Lee CB, Albertini MR, Flaherty LE, Petrella TM, Streicher H, Sondak VK, Kirkwood JM. Phase III Study of Adjuvant Ipilimumab (3 or 10 mg/kg) Versus High-Dose Interferon Alfa-2b for Resected High-Risk Melanoma: North American Intergroup E1609. J Clin Oncol 2020; 38:567-575. [PMID: 31880964 PMCID: PMC7030886 DOI: 10.1200/jco.19.01381] [Citation(s) in RCA: 125] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/06/2019] [Indexed: 12/28/2022] Open
Abstract
PURPOSE Phase III adjuvant trials have reported significant benefits in both relapse-free survival (RFS) and overall survival (OS) for high-dose interferon alfa (HDI) and ipilimumab at 10 mg/kg (ipi10). E1609 evaluated the safety and efficacy of ipilimumab at 3 mg/kg (ipi3) and ipi10 versus HDI. PATIENTS AND METHODS E1609 was a phase III trial in patients with resected cutaneous melanoma (American Joint Committee on Cancer 7th edition stage IIIB, IIIC, M1a, or M1b). It had 2 coprimary end points: OS and RFS. A 2-step hierarchic approach first evaluated ipi3 versus HDI followed by ipi10 versus HDI. RESULTS Between May 2011 and August 2014, 1,670 adult patients were centrally randomly assigned (1:1:1) to ipi3 (n = 523), HDI (n = 636), or ipi10 (n = 511). Treatment-related adverse events grade ≥ 3 occurred in 37% of patients receiving ipi3, 79% receiving HDI, and 58% receiving ipi10, with adverse events leading to treatment discontinuation in 35%, 20%, and 54%, respectively. Comparison of ipi3 versus HDI used an intent-to-treat analysis of concurrently randomly assigned patient cases (n = 1,051) and showed significant OS difference in favor of ipi3 (hazard ratio [HR], 0.78; 95.6% repeated CI, 0.61 to 0.99; P = .044; RFS: HR, 0.85; 99.4% CI, 0.66 to 1.09; P = .065). In the second step, for ipi10 versus HDI (n = 989), trends in favor of ipi10 did not achieve statistical significance. Salvage patterns after melanoma relapse showed significantly higher rates of ipilimumab and ipilimumab/anti-programmed death 1 use in the HDI arm versus ipi3 and ipi10 (P ≤ .001). CONCLUSION Adjuvant therapy with ipi3 benefits survival versus HDI; for the first time to our knowledge in melanoma adjuvant therapy, E1609 has demonstrated a significant improvement in OS against an active control regimen. The currently approved adjuvant ipilimumab dose (ipi10) was more toxic and not superior in efficacy to HDI.
Collapse
Affiliation(s)
| | - Sandra J. Lee
- Harvard Medical School, Boston, MA
- Dana-Farber Cancer Institute, Boston, MA
| | | | - Uma N. M. Rao
- University of Pittsburgh Medical Center, Pittsburgh, PA
| | | | - Omid Hamid
- Angeles Clinic & Research Institute, Santa Monica, CA
| | | | | | | | - Zeynep Eroglu
- H. Lee Moffitt Comprehensive Cancer Center, Tampa, FL
| | | | | | | | - David R. Minor
- Sutter-California Pacific Medical Center, San Francisco, CA
| | - Carrie B. Lee
- University of North Carolina at Chapel Hill, Chapel Hill, NC
| | | | | | | | | | | | | |
Collapse
|
165
|
Lin CY, Guu TW, Lai HC, Peng CY, Chiang JYJ, Chen HT, Li TC, Yang SY, Su KP, Chang JPC. Somatic pain associated with initiation of interferon-alpha (IFN-α) plus ribavirin (RBV) therapy in chronic HCV patients: A prospective study. Brain Behav Immun Health 2020; 2:100035. [PMID: 34589826 PMCID: PMC8474510 DOI: 10.1016/j.bbih.2019.100035] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 12/21/2019] [Accepted: 12/28/2019] [Indexed: 12/12/2022] Open
Abstract
Objective This study is aimed to investigate the association between interferon-alpha (IFN-α) plus ribavirin (RBV) treatment and emergence of somatic pain symptoms in patients with hepatitis C virus (HCV) over a 24-week treatment. Method In this prospective cohort study, 297 patients with HCV were evaluated at baseline and 2nd, 4th, 8th, 12th, 16th, 20th, and 24th week with structured Mini-International Neuropsychiatric Interview for Major Depressive Disorder (MDD) diagnosis and the Neurotoxicity Rating Scale (NRS) for somatic symptoms. Results Eighty-seven out of the 297 patients (29%) developed IFN-α induced depression and had significantly higher somatic pain symptoms as early as the 2nd week and at all the assessment time points (p < .001). Most depressed patients perceived greatest somatic pain at the 8th week of treatment. Moreover, NRS somatic pain scores after initial therapy strongly correlated with NRS somatic pain scores at all other assessment time points (p < .001). Conclusion IFN-α therapy induce significant somatic pain as early as the 2nd week of treatment in HCV patients who later developed MDD. Thus, initial NRS somatic pain score after initiation of IFN-α treatment may serve as a reference for the susceptibility of the individual to IFN-α induced depression.
Collapse
Affiliation(s)
- Chih Ying Lin
- School of Medicine, China Medical University (CMU), Taichung, Taiwan
- Mind-Body Interface Lab (MBI-Lab) and Department of Psychiatry, CMUH, Taichung, Taiwan
| | - Ta-Wei Guu
- Department Psychiatry, CMU Beigang Hospital, Yunlin, Taiwan
- Mind-Body Interface Lab (MBI-Lab) and Department of Psychiatry, CMUH, Taichung, Taiwan
| | - Hsueh-Chou Lai
- School of Medicine, China Medical University (CMU), Taichung, Taiwan
- Departement of Hepatogastroenterology, China Medial University Hospital, Taichung, Taiwan
| | - Cheng-Yuan Peng
- School of Medicine, China Medical University (CMU), Taichung, Taiwan
- Departement of Hepatogastroenterology, China Medial University Hospital, Taichung, Taiwan
| | - Jill Yi-Ju Chiang
- Mind-Body Interface Lab (MBI-Lab) and Department of Psychiatry, CMUH, Taichung, Taiwan
| | - Hui-Ting Chen
- Mind-Body Interface Lab (MBI-Lab) and Department of Psychiatry, CMUH, Taichung, Taiwan
| | - Tsai-Chung Li
- School of Medicine, China Medical University (CMU), Taichung, Taiwan
- China Medical University Graduate Institute of Biostatistics, Taiwan
| | - Shing-Yu Yang
- School of Medicine, China Medical University (CMU), Taichung, Taiwan
- China Medical University Graduate Institute of Biostatistics, Taiwan
| | - Kuan-Pin Su
- School of Medicine, China Medical University (CMU), Taichung, Taiwan
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
- Mind-Body Interface Lab (MBI-Lab) and Department of Psychiatry, CMUH, Taichung, Taiwan
| | - Jane Pei-Chen Chang
- School of Medicine, China Medical University (CMU), Taichung, Taiwan
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
- Mind-Body Interface Lab (MBI-Lab) and Department of Psychiatry, CMUH, Taichung, Taiwan
- Corresponding author. Department of Psychiatry, China Medical University Hospital, No. 2 Yu-Der Road, Taichung, 404, Taiwan.
| |
Collapse
|
166
|
Psioda MA, Ibrahim JG. Bayesian clinical trial design using historical data that inform the treatment effect. Biostatistics 2020; 20:400-415. [PMID: 29547966 DOI: 10.1093/biostatistics/kxy009] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 01/13/2018] [Indexed: 11/12/2022] Open
Abstract
We consider the problem of Bayesian sample size determination for a clinical trial in the presence of historical data that inform the treatment effect. Our broadly applicable, simulation-based methodology provides a framework for calibrating the informativeness of a prior while simultaneously identifying the minimum sample size required for a new trial such that the overall design has appropriate power to detect a non-null treatment effect and reasonable type I error control. We develop a comprehensive strategy for eliciting null and alternative sampling prior distributions which are used to define Bayesian generalizations of the traditional notions of type I error control and power. Bayesian type I error control requires that a weighted-average type I error rate not exceed a prespecified threshold. We develop a procedure for generating an appropriately sized Bayesian hypothesis test using a simple partial-borrowing power prior which summarizes the fraction of information borrowed from the historical trial. We present results from simulation studies that demonstrate that a hypothesis test procedure based on this simple power prior is as efficient as those based on more complicated meta-analytic priors, such as normalized power priors or robust mixture priors, when all are held to precise type I error control requirements. We demonstrate our methodology using a real data set to design a follow-up clinical trial with time-to-event endpoint for an investigational treatment in high-risk melanoma.
Collapse
Affiliation(s)
- Matthew A Psioda
- Department of Biostatistics, University of North Carolina, McGavran-Greenberg Hall, CB#7420, Chapel Hill, NC, USA
| | - Joseph G Ibrahim
- Department of Biostatistics, University of North Carolina, McGavran-Greenberg Hall, CB#7420, Chapel Hill, NC, USA
| |
Collapse
|
167
|
Gordy JT, Luo K, Kapoor A, Kim ES, Ayeh SK, Karakousis PC, Markham RB. Treatment with an immature dendritic cell-targeting vaccine supplemented with IFN-α and an inhibitor of DNA methylation markedly enhances survival in a murine melanoma model. Cancer Immunol Immunother 2020; 69:569-580. [PMID: 31980915 DOI: 10.1007/s00262-019-02471-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 12/31/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND The chemokine MIP-3α (CCL20) binds to CCR6 on immature dendritic cells. DNA vaccines fusing MIP-3α to melanoma-associated antigens have shown improved efficacy and immunogenicity in the B16F10 mouse melanoma model. Here, we report that the combination of type-I interferon therapy (IFNα) with 5-Aza-2'-deoxycitidine (5Aza) profoundly enhanced the therapeutic efficacy of a MIP-3α-Gp100-Trp2 DNA vaccine. METHODS Beginning on day 5 post-transplantation of B16F10 melanoma, vaccine was administered intramuscularly (i.m.) by electroporation. CpG adjuvant was given 2 days later. 5Aza was given intraperitoneally at 1 mg/kg and IFNα therapy either intratumorally or i.m. as noted. Tumor sizes, tumor growth, and mouse survival were assessed. Tumor lysate gene expression levels and tumor-infiltrating lymphocytes (TILs) were assessed by qRT-PCR and flow cytometry, respectively. RESULTS Adding IFNα and 5Aza treatments to mice vaccinated with MIP-3α-Gp100-Trp2 leads to reduced tumor burden and increased median survival (39% over vaccine and 95% over controls). Tumor lysate expression of CCL19 and CCR7 were upregulated ten and fivefold over vaccine, respectively. Vaccine-specific and overall CD8+ TILs were increased over vaccine (sevenfold and fourfold, respectively), as well as the proportion of TILs that were CD8+ (twofold). CONCLUSIONS Efficient targeting of antigen to immature dendritic cells with a chemokine-fusion vaccine offers an alternative to classic and dendritic cell vaccines. Combining this approach with IFNα and 5Aza treatment significantly improved vaccine efficacy. This improved efficacy correlated with changes in chemokine gene expression and CD8+ TIL infiltration and was dependent on the presence of all therapeutic components.
Collapse
Affiliation(s)
- James T Gordy
- The W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe Street, Baltimore, MD, 21205, USA
| | - Kun Luo
- The W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe Street, Baltimore, MD, 21205, USA
| | - Aakanksha Kapoor
- Department of Medicine, Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Emily S Kim
- Department of Medicine, Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Samuel K Ayeh
- Department of Medicine, Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Petros C Karakousis
- Department of Medicine, Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Richard B Markham
- The W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe Street, Baltimore, MD, 21205, USA.
| |
Collapse
|
168
|
Rahimi Kalateh Shah Mohammad G, Ghahremanloo A, Soltani A, Fathi E, Hashemy SI. Cytokines as potential combination agents with PD-1/PD-L1 blockade for cancer treatment. J Cell Physiol 2020; 235:5449-5460. [PMID: 31970790 DOI: 10.1002/jcp.29491] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Accepted: 01/08/2020] [Indexed: 12/11/2022]
Abstract
Immunotherapy has caused a paradigm shift in the treatment of several malignancies, particularly the blockade of programmed death-1 (PD-1) and its specific receptor/ligand PD-L1 that have revolutionized the treatment of a variety of malignancies, but significant durable responses only occur in a small percentage of patients, and other patients failed to respond to the treatment. Even those who initially respond can ultimately relapse despite maintenance treatment, there is considerable potential for synergistic combinations of immunotherapy and chemotherapy agents with immune checkpoint inhibitors into conventional cancer treatments. The clinical experience in the use of cytokines in the clinical setting indicated the efficiency of cytokine therapy in cancer immunotherapy. Combinational approaches to enhancing PD-L1/PD-1 pathways blockade efficacy with several cytokines such as interleukin (IL)-2, IL-15, IL-21, IL-12, IL-10, and interferon-α (IFN-α) may result in additional benefits. In this review, the current state of knowledge about PD-1/PD-L1 inhibitors, the date in the literature to ascertain the combination of anti-PD-1/PD-L1 antibodies with cytokines is discussed. Finally, it is noteworthy that novel therapeutic approaches based on the efficient combination of recombinant cytokines with the PD-L1/PD-1 blockade therapy can enhance antitumor immune responses against various malignancies.
Collapse
Affiliation(s)
| | - Atefeh Ghahremanloo
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Arash Soltani
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Esmat Fathi
- Department of Biological Sciences, University of Memphis, Memphis, Tennessee
| | - Seyed Isaac Hashemy
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Surgical Oncology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
169
|
Marinho ARS, Loschi RH. Bayesian cure fraction models with measurement error in the scale mixture of normal distribution. Stat Methods Med Res 2020; 29:2411-2444. [PMID: 31928318 DOI: 10.1177/0962280219893034] [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] [Indexed: 11/16/2022]
Abstract
Cure fraction models have been widely used to model time-to-event data when part of the individuals survives long-term after disease and are considered cured. Most cure fraction models neglect the measurement error that some covariates may experience which leads to poor estimates for the cure fraction. We introduce a Bayesian promotion time cure model that accounts for both mismeasured covariates and atypical measurement errors. This is attained by assuming a scale mixture of the normal distribution to describe the uncertainty about the measurement error. Extending previous works, we also assume that the measurement error variance is unknown and should be estimated. Three classes of prior distributions are assumed to model the uncertainty about the measurement error variance. Simulation studies are performed evaluating the proposed model in different scenarios and comparing it to the standard promotion time cure fraction model. Results show that the proposed models are competitive ones. The proposed model is fitted to analyze a dataset from a melanoma clinical trial assuming that the Breslow depth is mismeasured.
Collapse
Affiliation(s)
- Anna R S Marinho
- Departamento de Estatística, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Rosangela H Loschi
- Departamento de Estatística, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
170
|
Chu C, Liu S, Rong A. Study design of single-arm phase II immunotherapy trials with long-term survivors and random delayed treatment effect. Pharm Stat 2020; 19:358-369. [PMID: 31930622 DOI: 10.1002/pst.1976] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 08/28/2019] [Accepted: 09/09/2019] [Indexed: 01/25/2023]
Abstract
In the traditional study design of a single-arm phase II cancer clinical trial, the one-sample log-rank test has been frequently used. A common practice in sample size calculation is to assume that the event time in the new treatment follows exponential distribution. Such a study design may not be suitable for immunotherapy cancer trials, when both long-term survivors (or even cured patients from the disease) and delayed treatment effect are present, because exponential distribution is not appropriate to describe such data and consequently could lead to severely underpowered trial. In this research, we proposed a piecewise proportional hazards cure rate model with random delayed treatment effect to design single-arm phase II immunotherapy cancer trials. To improve test power, we proposed a new weighted one-sample log-rank test and provided a sample size calculation formula for designing trials. Our simulation study showed that the proposed log-rank test performs well and is robust of misspecified weight and the sample size calculation formula also performs well.
Collapse
Affiliation(s)
- Chenghao Chu
- Department of Biostatistics, Indiana University, Fairbanks School of Public Health, Indianapolis, IN, U.S.A
| | - Shufang Liu
- Data Science, Astellas Pharma Inc, Northbrook, IL, U.S.A
| | - Alan Rong
- Data Science, Astellas Pharma Inc, Northbrook, IL, U.S.A
| |
Collapse
|
171
|
Garbe C, Amaral T, Peris K, Hauschild A, Arenberger P, Bastholt L, Bataille V, Del Marmol V, Dréno B, Fargnoli MC, Grob JJ, Höller C, Kaufmann R, Lallas A, Lebbé C, Malvehy J, Middleton M, Moreno-Ramirez D, Pellacani G, Saiag P, Stratigos AJ, Vieira R, Zalaudek I, Eggermont AMM. European consensus-based interdisciplinary guideline for melanoma. Part 2: Treatment - Update 2019. Eur J Cancer 2019; 126:159-177. [PMID: 31866016 DOI: 10.1016/j.ejca.2019.11.015] [Citation(s) in RCA: 142] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 11/18/2019] [Indexed: 12/19/2022]
Abstract
A unique collaboration of multidisciplinary experts from the European Dermatology Forum, the European Association of Dermato-Oncology and the European Organization for Research and Treatment of Cancer (EORTC) was formed to make recommendations on cutaneous melanoma diagnosis and treatment, based on systematic literature reviews and the experts' experience. Cutaneous melanomas are excised with 1- to 2-cm safety margins. Sentinel lymph node dissection shall be performed as a staging procedure in patients with tumour thickness ≥1.0 mm or ≥0.8 mm with additional histological risk factors, although there is as yet no clear survival benefit for this approach. Therapeutic decisions in stage III/IV patients should be primarily made by an interdisciplinary oncology team ("Tumor Board"). Adjuvant therapies in stage III/IV patients are primarily anti-PD-1, independent of mutational status, or dabrafenib plus trametinib for BRAF-mutant patients. In distant metastasis, either resected or not, systemic treatment is indicated. For first-line treatment, particularly in BRAF wild-type patients, immunotherapy with PD-1 antibodies alone or in combination with CTLA-4 antibodies shall be considered. In particular scenarios for patients with stage IV melanoma and a BRAF-V600 E/K mutation, first-line therapy with BRAF/MEK inhibitors can be offered as an alternative to immunotherapy. In patients with primary resistance to immunotherapy and harbouring a BRAF-V600 E/K mutation, this therapy shall be offered in second-line. Systemic therapy in stage III/IV melanoma is a rapidly changing landscape, and it is likely that these recommendations may change in the near future.
Collapse
Affiliation(s)
- Claus Garbe
- Center for Dermatooncology, Department of Dermatology, Eberhard Karls University, Tuebingen, Germany.
| | - Teresa Amaral
- Center for Dermatooncology, Department of Dermatology, Eberhard Karls University, Tuebingen, Germany; Portuguese Air Force Health Care Direction, Lisbon, Portugal
| | - Ketty Peris
- Institute of Dermatology, Università Cattolica, Rome, Italy; Fondazione Policlinico Universitario A, Gemelli - IRCCS, Rome, Italy
| | - Axel Hauschild
- Department of Dermatology, University Hospital Schleswig-Holstein (UKSH), Campus Kiel, Kiel, Germany
| | - Petr Arenberger
- Department of Dermatovenerology, Third Faculty of Medicine, Charles University of Prague, Prague, Czech Republic
| | - Lars Bastholt
- Department of Oncology, Odense University Hospital, Denmark
| | - Veronique Bataille
- Twin Research and Genetic Epidemiology Unit, School of Basic & Medical Biosciences, King's College London, London, SE1 7EH, UK
| | - Veronique Del Marmol
- Department of Dermatology, Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Brigitte Dréno
- Dermatology Department, CHU Nantes, CIC 1413, CRCINA, University Nantes, Nantes, France
| | | | | | - Christoph Höller
- Department of Dermatology, Medical University of Vienna, Austria
| | - Roland Kaufmann
- Department of Dermatology, Venerology and Allergology, Frankfurt University Hospital, Frankfurt, Germany
| | - Aimilios Lallas
- First Department of Dermatology, Aristotle University, Thessaloniki, Greece
| | - Celeste Lebbé
- APHP Department of Dermatology, INSERM U976, University Paris 7 Diderot, Saint-Louis University Hospital, Paris, France
| | - Josep Malvehy
- Melanoma Unit, Department of Dermatology, Hospital Clinic, IDIBAPS, Barcelona, Spain
| | - Mark Middleton
- NIHR Biomedical Research Centre, University of Oxford, UK
| | - David Moreno-Ramirez
- Medical-&-Surgical Dermatology Service, Hospital Universitario Virgen Macarena, Sevilla, Spain
| | | | - Philippe Saiag
- University Department of Dermatology, Université de Versailles-Saint Quentin en Yvelines, APHP, Boulogne, France
| | - Alexander J Stratigos
- 1st Department of Dermatology, University of Athens School of Medicine, Andreas Sygros Hospital, Athens, Greece
| | - Ricardo Vieira
- Department of Dermatology and Venereology, Centro Hospitalar Universitário de Coimbra, Coimbra, Portugal
| | - Iris Zalaudek
- Dermatology Clinic, Maggiore Hospital, University of Trieste, Trieste, Italy
| | | |
Collapse
|
172
|
Stellato D, Thabane M, Eichten C, Delea TE. Preferences of Canadian patients and physicians for adjuvant treatments for melanoma. ACTA ACUST UNITED AC 2019; 26:e755-e765. [PMID: 31896946 DOI: 10.3747/co.26.5085] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background Past research suggests that patients with early- and late-stage melanoma will endure adverse events and inconvenient treatment regimens for improved survival. Evidence about the preferences of Canadian patients and physicians for novel adjuvant treatments for melanoma is unavailable. Methods Patient and physician preferences for adjuvant treatments for melanoma were assessed in an online discrete choice experiment (dce). Treatment alternatives were characterized by 8 attributes with respect to dosing regimen, efficacy, and toxicities, with levels corresponding to those for dabrafenib-trametinib, nivolumab, pembrolizumab, and interferon. For patients, the effects of melanoma on quality of life and ability to work and perform activities of daily living were also assessed. Patients were recruited by Canadian melanoma patient advocacy groups through e-mail and social media. Physicians were recruited by e-mail. Results Of 94 patients who started the survey, 51 completed 1 or more dce questions. Of 166 physicians sent the e-mail invitation, 18 completed 1 or more dce questions. For patients, an increased probability of remaining cancer-free over 21 months was the most important attribute. For physicians, an increased chance of the patient's remaining alive over 36 months was the most important attribute. Patients and physicians chose active treatment over no treatment 85% and 86% of the time respectively and a treatment with attributes consistent with dabrafenib-trametinib 71% and 67% of the time respectively. A substantial proportion of patients reported worrying about future diagnostic tests and their cancer coming back. Conclusions Canadian patients and physicians are generally concordant in their preferences for adjuvant melanoma treatments, preferring active treatment to no treatment and dabrafenib-trametinib to other options.
Collapse
Affiliation(s)
- D Stellato
- Policy Analysis Inc., Brookline, MA, U.S.A
| | - M Thabane
- Novartis Pharmaceuticals Canada, Dorval, QC
| | - C Eichten
- Policy Analysis Inc., Brookline, MA, U.S.A
| | - T E Delea
- Policy Analysis Inc., Brookline, MA, U.S.A
| |
Collapse
|
173
|
Testori AAE, Ribero S, Indini A, Mandalà M. Adjuvant Treatment of Melanoma: Recent Developments and Future Perspectives. Am J Clin Dermatol 2019; 20:817-827. [PMID: 31177507 DOI: 10.1007/s40257-019-00456-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Surgical excision is the treatment of choice for early stage melanoma, and this strategy is initially curative for the vast majority of patients. However, only approximately 40-60% of high-risk patients who undergo surgery alone will be disease-free at 5 years. These patients will ultimately experience loco-regional relapse or relapse at distant sites. The main aim of adjuvant therapies is to reduce the recurrence rate of radically operated patients at high risk and to potentially improve survival. Recent practice changing results with immune checkpoint inhibitors and targeted therapies have been published in stage III/IV melanoma patients, after surgical complete resection, and have dramatically improved the landscape of adjuvant therapy. Interferon-α, ipilimumab, and more recently anti-programmed cell death protein-1 antibodies and BRAF inhibitors plus MEK inhibitors have been approved in the adjuvant setting by the US Food and Drug Administration; similarly, the same drugs are approved by the European Medicines Agency with the exception of ipilimumab. A completely new scenario is emerging in the neoadjuvant setting as well: in locally advanced or metastatic disease, patients may partially respond to neoadjuvant therapy and become virtually resectable with systemic control of disease. This review summarizes the current state of the field and describes new strategies tracing the history of adjuvant therapy in melanoma, with a view on future projects.
Collapse
Affiliation(s)
| | - Simone Ribero
- Medical Sciences Department, Dermatologic Clinic, University of Turin, Turin, Italy
| | - Alice Indini
- Melanoma Unit, Department of Oncology and Hematology, Papa Giovanni XXIII Hospital, Bergamo, Italy
| | - Mario Mandalà
- Melanoma Unit, Department of Oncology and Hematology, Papa Giovanni XXIII Hospital, Bergamo, Italy
| |
Collapse
|
174
|
Sharma R, Koruth R, Kanters S, Druyts E, Tarhini A. Comparative efficacy and safety of dabrafenib in combination with trametinib versus competing adjuvant therapies for high-risk melanoma. J Comp Eff Res 2019; 8:1349-1363. [PMID: 31778073 DOI: 10.2217/cer-2019-0061] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim To conduct a systematic literature review of high-risk resectable cutaneous melanoma adjuvant therapeutics and compare safety and efficacy. Methods: The systematic literature review included randomized controlled trials investigating: dabrafenib plus trametinib (DAB + TRAM), nivolumab, pembrolizumab, ipilimumab, vemurafenib, chemotherapy and interferons. Outcomes included overall survival (OS), relapse-free survival, distant metastasis-free survival and safety. All outcomes were synthesized using Bayesian network meta-analysis. Results: Across relapse-free survival, distant metastasis-free survival and OS, DAB + TRAM had the lowest estimated hazards of respective events relative to all other treatments (exception relative to nivolumab in OS). Differences were significant relative to placebo, chemotherapy, interferons and ipilimumab. Conclusion: DAB + TRAM has improved efficacy over historical treatment options (ipilimumab, interferons and chemotherapy) and comparable efficacy with other targeted and immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Rohini Sharma
- Precision Xtract, Vancouver, British Columbia, V6H 3Y4, Canada
| | - Roy Koruth
- Novartis Pharmaceuticals Corporation, East Hanover, NJ 07936, USA
| | - Steve Kanters
- Precision Xtract, Vancouver, British Columbia, V6H 3Y4, Canada
| | - Eric Druyts
- Precision Xtract, Vancouver, British Columbia, V6H 3Y4, Canada
- Novartis Pharmaceuticals Corporation, East Hanover, NJ 07936, USA
- Moffitt Comprehensive Cancer Center, Tampa, FL 33612, USA
| | - Ahmad Tarhini
- Moffitt Comprehensive Cancer Center, Tampa, FL 33612, USA
| |
Collapse
|
175
|
Fayne RA, Macedo FI, Rodgers SE, Möller MG. Evolving management of positive regional lymph nodes in melanoma: Past, present and future directions. Oncol Rev 2019; 13:433. [PMID: 31857858 PMCID: PMC6902307 DOI: 10.4081/oncol.2019.433] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 11/20/2019] [Indexed: 12/29/2022] Open
Abstract
Sentinel lymph node (SLN) biopsy has become the standard of care for lymph node staging in melanoma and the most important predictor of survival in clinically node-negative disease. Previous guidelines recommend completion lymph node dissection (CLND) in cases of positive SLN; however, the lymph nodes recovered during CLND are only positive in a minority of these cases. Recent evidence suggests that conservative management (i.e. observation) has similar outcomes compared to CLND. We sought to review the most current literature regarding the management of SLN in metastatic melanoma and to discuss potential future directions.
Collapse
Affiliation(s)
- Rachel A Fayne
- Division of Surgical Oncology, Dewitt-Daughtry Department of Surgery, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine
| | - Francisco I Macedo
- Department of Surgery, North Florida Regional Medical Center, University of Central Florida College of Medicine, Miami, FL, USA
| | - Steven E Rodgers
- Division of Surgical Oncology, Dewitt-Daughtry Department of Surgery, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine
| | - Mecker G Möller
- Division of Surgical Oncology, Dewitt-Daughtry Department of Surgery, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine
| |
Collapse
|
176
|
|
177
|
Samuel E, Moore M, Voskoboynik M, Shackleton M, Haydon A. An update on adjuvant systemic therapies in melanoma. Melanoma Manag 2019; 6:MMT28. [PMID: 31807279 PMCID: PMC6891936 DOI: 10.2217/mmt-2019-0009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
There is a global increase in the incidence of melanoma, with approximately 300,000 new cases in 2018 worldwide, according to statistics from the International Agency for Research on Cancer. With this rising incidence, it is important to optimize treatment strategies in all stages of the disease to provide better patient outcomes. The role of adjuvant therapy in patients with resected stage 3 melanoma is a rapidly evolving field. Interferon was the first agent shown to have any utility in this space, however, recent advances in both targeted therapies and immunotherapies have led to a number of practice changing adjuvant trials in resected stage 3 disease.
Collapse
Affiliation(s)
- Evangeline Samuel
- Department of Medical Oncology, Monash Health, Clayton, Melbourne 3168, Australia
| | - Maggie Moore
- Department of Medical Oncology, The Alfred Hospital, Melbourne 3004, Australia
| | - Mark Voskoboynik
- Department of Medical Oncology, The Alfred Hospital, Melbourne 3004, Australia
| | - Mark Shackleton
- Department of Medical Oncology, The Alfred Hospital, Melbourne 3004, Australia
| | - Andrew Haydon
- Department of Medical Oncology, The Alfred Hospital, Melbourne 3004, Australia
| |
Collapse
|
178
|
Abstract
Over the past decade, preclinical and clinical research have confirmed the essential role of interferons for effective host immunological responses to malignant cells. Type I interferons (IFNα and IFNβ) directly regulate transcription of >100 downstream genes, which results in a myriad of direct (on cancer cells) and indirect (through immune effector cells and vasculature) effects on the tumour. New insights into endogenous and exogenous activation of type I interferons in the tumour and its microenvironment have given impetus to drug discovery and patient evaluation of interferon-directed strategies. When combined with prior observations or with other effective modalities for cancer treatment, modulation of the interferon system could contribute to further reductions in cancer morbidity and mortality. This Review discusses new interferon-directed therapeutic opportunities, ranging from cyclic dinucleotides to genome methylation inhibitors, angiogenesis inhibitors, chemoradiation, complexes with neoantigen-targeted monoclonal antibodies, combinations with other emerging therapeutic interventions and associations of interferon-stimulated gene expression with patient prognosis - all of which are strategies that have or will soon enter translational clinical evaluation.
Collapse
|
179
|
Lamberti MJ, Mentucci FM, Roselli E, Araya P, Rivarola VA, Rumie Vittar NB, Maccioni M. Photodynamic Modulation of Type 1 Interferon Pathway on Melanoma Cells Promotes Dendritic Cell Activation. Front Immunol 2019; 10:2614. [PMID: 31781113 PMCID: PMC6856948 DOI: 10.3389/fimmu.2019.02614] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Accepted: 10/21/2019] [Indexed: 01/21/2023] Open
Abstract
The immune response against cancer generated by type-I-interferons (IFN-1) has recently been described. Exogenous and endogenous IFN-α/β have an important role in immune surveillance and control of tumor development. In addition, IFN-1s have recently emerged as novel DAMPs for the consecutive events connecting innate and adaptive immunity, and they also have been postulated as an essential requirement for induction of immunogenic cell death (ICD). In this context, photodynamic therapy (PDT) has been previously linked to the ICD. PDT consists in the administration of a photosensitizer (PS) and its activation by irradiation of the affected area with visible light producing excitation of the PS. This leads to the local generation of harmful reactive oxygen species (ROS) with limited or no systemic defects. In the current work, Me-ALA inducing PpIX (endogenous PS) was administrated to B16-OVA melanoma cells. PpIX preferentially localized in the endoplasmic reticulum (ER). Subsequent PpIX activation with visible light significantly induced oxidative ER-stress mediated-apoptotic cell death. Under these conditions, the present study was the first to report the in vitro upregulation of IFN-1 expression in response to photodynamic treatment in melanoma. This IFN-α/β transcripts upregulation was concurrent with IRF-3 phosphorylation at levels that efficiently activated STAT1 and increased ligand receptor (cGAS) and ISG (CXCL10, MX1, ISG15) expression. The IFN-1 pathway has been identified as a critical molecular pathway for the antitumor host immune response, more specifically for the dendritic cells (DCs) functions. In this sense, PDT-treated melanoma cells induced IFN-1-dependent phenotypic maturation of monocyte-derived dendritic cells (DCs) by enhancing co-stimulatory signals (CD80, MHC-II) and tumor-directed chemotaxis. Collectively, our findings showed a new effect of PDT-treated cancer cells by modulating the IFN-1 pathway and its impact on the activation of DCs, emphasizing the potential relevance of PDT in adoptive immunotherapy protocols.
Collapse
Affiliation(s)
- María Julia Lamberti
- Departamento de Biología Molecular, Facultad de Ciencias Exactas, Físico-Químicas y Naturales, Instituto de Biotecnología Ambiental y Salud, Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad Nacional de Río Cuarto, Córdoba, Argentina
| | - Fátima María Mentucci
- Departamento de Biología Molecular, Facultad de Ciencias Exactas, Físico-Químicas y Naturales, Instituto de Biotecnología Ambiental y Salud, Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad Nacional de Río Cuarto, Córdoba, Argentina
| | - Emiliano Roselli
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Centro de Investigaciones en Bioquímica Clínica e Inmunología, Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Paula Araya
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Centro de Investigaciones en Bioquímica Clínica e Inmunología, Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Viviana Alicia Rivarola
- Departamento de Biología Molecular, Facultad de Ciencias Exactas, Físico-Químicas y Naturales, Instituto de Biotecnología Ambiental y Salud, Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad Nacional de Río Cuarto, Córdoba, Argentina
| | - Natalia Belén Rumie Vittar
- Departamento de Biología Molecular, Facultad de Ciencias Exactas, Físico-Químicas y Naturales, Instituto de Biotecnología Ambiental y Salud, Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad Nacional de Río Cuarto, Córdoba, Argentina
| | - Mariana Maccioni
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Centro de Investigaciones en Bioquímica Clínica e Inmunología, Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| |
Collapse
|
180
|
Gamboa AC, Lowe M, Yushak ML, Delman KA. Surgical Considerations and Systemic Therapy of Melanoma. Surg Clin North Am 2019; 100:141-159. [PMID: 31753109 DOI: 10.1016/j.suc.2019.09.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Recent advances in effective medical therapies have markedly improved the prognosis for patients with advanced melanoma. This article aims to highlight the current era of integrated multidisciplinary care of patients with advanced melanoma by outlining current approved therapies, including immunotherapy, targeted therapy, radiation therapy, and other strategies used in both the adjuvant and the neoadjuvant setting as well as the evolving role of surgical intervention in the changing landscape of advanced melanoma.
Collapse
Affiliation(s)
- Adriana C Gamboa
- Division of Surgical Oncology, Emory University School of Medicine, 1365B Clifton Road Northeast, Suite B4000, Atlanta, GA 30322, USA
| | - Michael Lowe
- Division of Surgical Oncology, Emory University School of Medicine, 1365B Clifton Road Northeast, Suite B4000, Atlanta, GA 30322, USA
| | - Melinda L Yushak
- Division of Medical Oncology, Emory University School of Medicine, 1365B4 Clifton Road Northeast, Suite B4000, Atlanta, GA 30322, USA
| | - Keith A Delman
- Division of Surgical Oncology, Emory University School of Medicine, 1365B Clifton Road Northeast, Suite B4000, Atlanta, GA 30322, USA.
| |
Collapse
|
181
|
Abstract
Advanced/metastatic melanoma is an aggressive cancer with a low survival rate. Traditional cytotoxic chemotherapies do not appreciably extend life and systemic cytokine/chemokine administration produces toxic side effects. By harnessing the surveillance and cytotoxic features of the immune system, immunotherapies can provide a durable response and are proved to improve disease outcomes in patients with advanced/metastatic melanoma and other cancers. Close monitoring is necessary, however, to identify and treat immune system-related adverse events before they become life-threatening. Because metastatic lesions can respond differently to immunotherapies, modified response criteria have been developed to assist physicians in tracking patient response to treatment.
Collapse
Affiliation(s)
- Adedayo A Onitilo
- Department of Hematology/Oncology, Marshfield Clinic - Weston Center, 3501 Cranberry Boulevard, Weston, WI 54476, USA.
| | - Jaimie A Wittig
- Pharmacy Services, Marshfield Medical Center, 1000 North Oak Avenue, Marshfield, WI 54449, USA
| |
Collapse
|
182
|
Stellato D, Gerbasi ME, Ndife B, Ghate SR, Moynahan A, Mishra D, Gunda P, Koruth R, Delea TE. Budget Impact of Dabrafenib and Trametinib in Combination as Adjuvant Treatment of BRAF V600E/K Mutation-Positive Melanoma from a U.S. Commercial Payer Perspective. J Manag Care Spec Pharm 2019; 25:1227-1237. [PMID: 31663466 PMCID: PMC10398148 DOI: 10.18553/jmcp.2019.25.11.1227] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Before the approval of dabrafenib and trametinib in combination, there were no approved therapies in the adjuvant setting that target the RAS/RAF/MEK/ERK pathway. OBJECTIVE To evaluate the budget impact of dabrafenib and trametinib in combination for adjuvant treatment of patients with BRAF V600 mutation-positive resected Stage IIIA, IIIB, or IIIC melanoma from a U.S. commercial payer perspective using data from the COMBI-AD trial, as well as other sources. METHODS The budget impact of dabrafenib and trametinib in combination for patients with BRAF V600E/K mutation-positive, resected Stage IIIA, IIIB, or IIIC melanoma was evaluated from the perspective of a hypothetical population of 1 million members with demographic characteristics consistent with those of a commercially insured U.S. insurance plan (i.e., adults aged less than 65 years) using an economic model developed in Microsoft Excel. The model compared melanoma-related health care costs over a 3-year projection period under 2 scenarios: (1) a reference scenario in which dabrafenib and trametinib are assumed to be unavailable for adjuvant therapy and (2) a new scenario in which the combination is assumed to be available. Treatments potentially displaced by dabrafenib and trametinib were assumed to include observation, high-dose interferon alpha-2b, ipilimumab, and nivolumab. Costs considered in the model include those of adjuvant therapies and treatment of locoregional and distant recurrences. The numbers of patients eligible for treatment with dabrafenib and trametinib were based on data from cancer registries, published sources, and assumptions. Treatment mixes under the reference and new scenarios were based on market research data, clinical expert opinion, and assumptions. Probabilities of recurrence and death were based on data from the COMBI-AD trial and an indirect treatment comparison. Medication costs were based on wholesale acquisition cost prices. Costs of distant recurrence were from a health insurance claims study. RESULTS In a hypothetical population of 1 million commercially insured members, 48 patients were estimated to become eligible for treatment with dabrafenib and trametinib in combination over the 3-year projection period; in the new scenario, 10 patients were projected to receive such treatment. Cumulative costs of melanoma-related care were estimated to be $6.3 million in the reference scenario and $6.9 million in the new scenario. The budget impact of dabrafenib and trametinib in combination was an increase of $549 thousand overall and 1.5 cents per member per month. CONCLUSIONS For a hypothetical U.S. commercial health plan of 1 million members, the budget impact of dabrafenib and trametinib in combination as adjuvant treatment for melanoma is likely to be relatively modest and within the range of published estimates for oncology therapies. These results may assist payers in making coverage decisions regarding the use of adjuvant dabrafenib and trametinib in melanoma. DISCLOSURES Funding for this research was provided to Policy Analysis Inc. (PAI) by Novartis Pharmaceuticals. Stellato, Moynahan, and Delea are employed by PAI. Ndife, Koruth, Mishra, and Gunda are employed by Novartis. Ghate was employed by Novartis at the time of this study and is shareholder in Novartis, Provectus Biopharmaceuticals, and Mannkind Corporation. Gerbasi was employed by PAI at the time of this study and is currently an employee, and stockholder, of Sage Therapeutics. Delea reports grant funding from Merck and research funding from Amgen, Novartis, Sanofi, Seattle Genetics, Takeda, Jazz, EMD Serono, and 21st Century Oncology, unrelated to this work.
Collapse
Affiliation(s)
| | | | - Briana Ndife
- Novartis Pharmaceuticals, East Hanover, New Jersey
| | | | | | - Dinesh Mishra
- Novartis Pharmaceuticals, Hyderabad, Telangana, India
| | - Praveen Gunda
- Novartis Pharmaceuticals, Hyderabad, Telangana, India
| | - Roy Koruth
- Novartis Pharmaceuticals, East Hanover, New Jersey
| | | |
Collapse
|
183
|
Survival Comparison between Melanoma Patients Treated with Patient-Specific Dendritic Cell Vaccines and Other Immunotherapies Based on Extent of Disease at the Time of Treatment. Biomedicines 2019; 7:biomedicines7040080. [PMID: 31614482 PMCID: PMC6966441 DOI: 10.3390/biomedicines7040080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 10/01/2019] [Accepted: 10/02/2019] [Indexed: 11/16/2022] Open
Abstract
Encouraging survival was observed in single arm and randomized phase 2 trials of patient-specific dendritic cell vaccines presenting autologous tumor antigens from autologous cancer cells that were derived from surgically resected metastases whose cells were self-renewing in vitro. Based on most advanced clinical stage and extent of tumor at the time of treatment, survival was best in patients classified as recurrent stage 3 without measurable disease. Next best was in stage 4 without measurable disease, and the worst survival was for measurable stage 4 disease. In this study, the survival of these patients was compared to the best contemporary controls that were gleaned from the clinical trial literature. The most comparable controls typically were from clinical trials testing other immunotherapy approaches. Even though contemporary controls typically had better prognostic features, median and/or long-term survival was consistently better in patients treated with this dendritic cell vaccine, except when compared to anti-programmed death molecule 1 (anti-PD-1). The clinical benefit of this patient-specific vaccine appears superior to a number of other immunotherapy approaches, but it is more complex to deliver than anti-PD-1 while equally effective. However, there is a strong rationale for combining such a product with anti-PD-1 in the treatment of patients with metastatic melanoma.
Collapse
|
184
|
Mohr P, Kiecker F, Soriano V, Dereure O, Mujika K, Saiag P, Utikal J, Koneru R, Robert C, Cuadros F, Chacón M, Villarroel RU, Najjar YG, Kottschade L, Couselo EM, Koruth R, Guérin A, Burne R, Ionescu-Ittu R, Perrinjaquet M, Zager JS. Adjuvant therapy versus watch-and-wait post surgery for stage III melanoma: a multicountry retrospective chart review. Melanoma Manag 2019; 6:MMT33. [PMID: 31871622 PMCID: PMC6923782 DOI: 10.2217/mmt-2019-0015] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 09/11/2019] [Indexed: 12/11/2022] Open
Abstract
AIM To describe treatment patterns among patients with stage III melanoma who underwent surgical excision in years 2011-2016, and assess outcomes among patients who subsequently received systemic adjuvant therapy versus watch-and-wait. METHODS Chart review of 380 patients from 17 melanoma centers in North America, South America and Europe. RESULTS Of 129 (34%) patients treated with adjuvant therapy, 85% received interferon α-2b and 56% discontinued treatment (mostly due to adverse events). Relapse-free survival was significantly longer for patients treated with adjuvant therapy versus watch-and-wait (hazard ratio = 0.63; p < 0.05). There was considerable heterogeneity in adjuvant treatment schedules and doses. Similar results were found in patients who received interferon-based adjuvant therapy. CONCLUSION Adjuvant therapies with better safety/efficacy profiles will improve clinical outcomes in patients with stage III melanoma.
Collapse
Affiliation(s)
- Peter Mohr
- Department of Dermatology, Elbe Kliniken, Stade, Germany
| | - Felix Kiecker
- Department of Dermatology and Allergy, Skin Cancer Center, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Virtudes Soriano
- Department of Medical Oncology, Fundación Instituto Valenciano de Oncología, Valencia, Spain
| | - Olivier Dereure
- Department of Dermatology and INSERM U1058 ‘pathogenesis and control of chronic infections’, University of Montpellier, Montpellier, France
| | - Karmele Mujika
- Department of Medical Oncology, Onkologikoa-Oncology Institute Gipuzkoa, Gipuzkoa, Spain
| | - Philippe Saiag
- Department of General and Oncologic Dermatology Ambroise Paré Hospital, APHP; EA 4340 ‘Biomarkers in cancerology and hemato-oncology’, UVSQ, Université Paris-Saclay, Boulogne-Billancourt, France
| | - Jochen Utikal
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany and Department of Dermatology, Venereology and Allergology; University Medical Center, Ruprecht-Karl University of Heidelberg, Mannheim, Germany
| | - Rama Koneru
- RS McLaughlin Durham Regional Cancer Centre, Lakeridge Health, Oshawa, Oshawa, Ontario, Canada
| | - Caroline Robert
- Dermatology Unit, Gustave Roussy and Paris-Saclay University, Villejuif, France
| | - Florencia Cuadros
- Medical Oncology, Instituto de Oncologia de Rosario, Rosario, Santa Fe, Argentina
| | - Matias Chacón
- Departments of Medical and Surgical Oncology, Instituto Alexander Fleming, Buenos Aires, Argentina
| | | | - Yana G Najjar
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Lisa Kottschade
- Department of Medical Oncology, Mayo Clinic, Rochester, MN 55905, USA
| | - Eva M Couselo
- Department of Medical Oncology, Vall d'Hebron Hospital and VHIO (Vall d'Hebron Institute of Oncology), Barcelona, Spain
| | - Roy Koruth
- Novartis Pharmaceuticals Corporation, East Hanover, NJ 07936, USA
| | | | | | | | | | - Jonathan S Zager
- Departments of Cutaneous Oncology and Sarcoma, Moffitt Cancer Center, Tampa, FL 33612, USA
| |
Collapse
|
185
|
Zibelman M, Plimack ER. Pembrolizumab plus ipilimumab or pegylated interferon alfa-2b for patients with melanoma or renal cell carcinoma: take new drugs but keep the old? ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:S95. [PMID: 31576303 DOI: 10.21037/atm.2019.04.57] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Matthew Zibelman
- Department of Hematology/Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Elizabeth R Plimack
- Department of Hematology/Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA
| |
Collapse
|
186
|
Egger ME, Scoggins CR, McMasters KM. The Sunbelt Melanoma Trial. Ann Surg Oncol 2019; 27:28-34. [PMID: 31529312 DOI: 10.1245/s10434-019-07828-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Indexed: 12/11/2022]
Abstract
The Sunbelt Melanoma Trial, a multicenter, prospective randomized clinical study, evaluated the role of high-dose interferon alfa-2b (HDI) therapy for patients with a single positive sentinel lymph node (SLN) metastasis treated with a completion lymph node dissection (CLND). A second protocol in the trial evaluated the prognostic significance of using molecular markers to identify submicroscopic metastases in sentinel lymph nodes that were negative by routine pathologic analysis. The role of CLND with or without adjuvant HDI was evaluated in this group of patients. The results of the study demonstrated that adjuvant HDI offered no survival benefit for patients with a single positive SLN in terms of disease-free or overall survival. Molecular staging using polymerase chain reaction (PCR) for melanoma markers did not identify a high-risk group of patients at increased risk of melanoma recurrence. Additional treatment of these patients who were PCR-positive with either CLND alone or CLND plus HDI did not improve their survival. Additional studies from the Sunbelt Melanoma Trial helped to validate the operational standards of the SLN biopsy procedure and defined the complication rates for both SLN biopsy and CLND. A prognostic risk calculator has been developed from trial data, and the importance of different micrometastatic tumor burden measurements was reported. Although the Sunbelt Melanoma Trial did not demonstrate an improvement in survival with HDI, it is an important trial that highlights the significance of surgeon-initiated randomized clinical trials that incorporate surgical techniques, molecular biomarkers, and adjuvant therapy.
Collapse
Affiliation(s)
- Michael E Egger
- The Hiram C Polk, Jr, MD Department of Surgery, University of Louisville School of Medicine, Louisville, KY, USA.
| | - Charles R Scoggins
- The Hiram C Polk, Jr, MD Department of Surgery, University of Louisville School of Medicine, Louisville, KY, USA
| | - Kelly M McMasters
- The Hiram C Polk, Jr, MD Department of Surgery, University of Louisville School of Medicine, Louisville, KY, USA
| |
Collapse
|
187
|
Najjar YG, Puligandla M, Lee SJ, Kirkwood JM. An updated analysis of 4 randomized ECOG trials of high-dose interferon in the adjuvant treatment of melanoma. Cancer 2019; 125:3013-3024. [PMID: 31067358 PMCID: PMC7428054 DOI: 10.1002/cncr.32162] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 03/04/2019] [Accepted: 03/11/2019] [Indexed: 11/05/2022]
Abstract
BACKGROUND The pivotal E1684, E1690, E1694, and E2696 trials of adjuvant high-dose interferon-α (HDI) enrolled nearly 2000 patients, and established HDI as the standard of care in adjuvant therapy for patients with resected high-risk melanoma. Herein, the authors present an updated analysis of these 4 trials. METHODS Survival and disease status were updated in September 2016. These data represent a median follow-up of 17.9 years for the E1684 trial, 12.2 years for the E1690 trial, 16.0 years for the E1694 trial, and 16.5 years for the E2696 trial. RESULTS The current analysis confirmed the benefit to recurrence-free survival (RFS) of HDI in the E1684 trial at a median follow-up of 17.9 years. The RFS benefit in the E1694 trial remained evident at a median follow-up of 16 years. Furthermore, the results of the current study confirmed the RFS benefit of adjuvant HDI compared with observation in a pooled analysis of the E1684 and E1690 trials. No overall survival benefit was apparent in this pooled analysis. Updated results for the E1690 and E2696 trials did not differ from those previously reported. In addition, to the authors' knowledge, the current study is the first to report a significant difference in melanoma-specific survival (MSS) between patients treated with HDI compared with the ganglioside GM2/keyhole limpet hemocyanin (GMK) vaccine in the E1694 trial. CONCLUSIONS In patients with resected high-risk melanoma, adjuvant HDI demonstrated improved RFS in the E1684 and E1694 trials, and improved MSS in a pooled analysis of HDI in the E1694 trial. To the authors' knowledge, these findings represent the most mature level of evidence for the benefit of HDI with respect to RFS and MSS. HDI is the only approved adjuvant treatment for which there are data available in patients with resected stage IIB/IIC melanoma, and remains a reasonable treatment option in this population.
Collapse
Affiliation(s)
- Yana G. Najjar
- Department of Medicine, Division of Hematology-Oncology, University of Pittsburgh, UPMC-Hillman Cancer Center. 5117 Centre Ave, 1.32 E, Pittsburgh, PA 15213
| | - Maneka Puligandla
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute
| | - Sandra J. Lee
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Harvard Medical School
| | - John M. Kirkwood
- Department of Medicine, Division of Hematology-Oncology, University of Pittsburgh, UPMC-Hillman Cancer Center
| |
Collapse
|
188
|
Weiss SA, Wolchok JD, Sznol M. Immunotherapy of Melanoma: Facts and Hopes. Clin Cancer Res 2019; 25:5191-5201. [PMID: 30923036 PMCID: PMC6726509 DOI: 10.1158/1078-0432.ccr-18-1550] [Citation(s) in RCA: 208] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 02/21/2019] [Accepted: 03/25/2019] [Indexed: 12/14/2022]
Abstract
Melanoma is among the most sensitive of malignancies to immune modulation. Although multiple trials conducted over decades with vaccines, cytokines, and cell therapies demonstrated meaningful responses in a small subset of patients with metastatic disease, a true increase in overall survival (OS) within a randomized phase III trial was not observed until the development of anti-CTLA-4 (ipilimumab). Further improvements in OS for metastatic disease were observed with the anti-PD-1-based therapies (nivolumab, pembrolizumab) as single agents or combined with ipilimumab. A lower bound for expected 5-year survival for metastatic melanoma is currently approximately 35% and could be as high as 50% for the nivolumab/ipilimumab combination among patients who would meet criteria for clinical trials. Moreover, a substantial fraction of long-term survivors will likely remain progression-free without continued treatment. The hope and major challenge for the future is to understand the immunobiology of tumors with primary or acquired resistance to anti-PD-1 or anti-PD-1/anti-CTLA-4 and to develop effective immune therapies tailored to individual patient subsets not achieving long-term clinical benefit. Additional goals include optimal integration of immune therapy with nonimmune therapies, the development and validation of predictive biomarkers in the metastatic setting, improved prognostic and predictive biomarkers for the adjuvant setting, understanding mechanisms of and decreasing toxicity, and optimizing the duration of therapy.
Collapse
Affiliation(s)
- Sarah A Weiss
- Yale University School of Medicine, New Haven, Connecticut.
| | - Jedd D Wolchok
- Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, New York
| | - Mario Sznol
- Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
189
|
Ollila DW, Meyers MO. Time may Heal All Wounds, but While It Does, Melanoma Marches on. Ann Surg Oncol 2019; 26:3800-3802. [PMID: 31468216 DOI: 10.1245/s10434-019-07674-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Indexed: 11/18/2022]
Affiliation(s)
- David W Ollila
- Department of Surgery, Division of Surgical Oncology, University of North Carolina, Chapel Hill, NC, USA.
| | - Michael O Meyers
- Department of Surgery, Division of Surgical Oncology, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
190
|
Spagnolo F, Boutros A, Tanda E, Queirolo P. The adjuvant treatment revolution for high-risk melanoma patients. Semin Cancer Biol 2019; 59:283-289. [PMID: 31445219 DOI: 10.1016/j.semcancer.2019.08.024] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 08/05/2019] [Accepted: 08/20/2019] [Indexed: 01/06/2023]
Abstract
The past 5 years have witnessed the results of many practice-changing studies that have dramatically improved the landscape of adjuvant therapy in patients with resected, high-risk melanoma. After a 20-year era of adjuvant interferon, the anti-CTLA-4 and anti-PD-1 immune-checkpoint inhibitors, and MAPK-directed targeted therapy brought a revolution into the adjuvant treatment of melanoma. These results came along with the practice-changing results of two large multicenter studies showing no benefit in terms of overall survival for completion lymph node dissection after positive sentinel node biopsy. In this review, we summarized the current state of the art of the adjuvant treatment of high-risk melanoma, with a view on future perspectives.
Collapse
Affiliation(s)
| | - Andrea Boutros
- Skin Cancer Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Enrica Tanda
- Skin Cancer Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Paola Queirolo
- Division of Medical Oncology for Melanoma, Sarcoma, and Rare Tumors, IEO, European Institute of Oncology IRCCS, Milan, Italy.
| |
Collapse
|
191
|
Umemoto S, Haruta M, Sakisaka M, Ikeda T, Tsukamoto H, Komohara Y, Takeya M, Nishimura Y, Senju S. Cancer therapy with major histocompatibility complex-deficient and interferon β-producing myeloid cells derived from allogeneic embryonic stem cells. Cancer Sci 2019; 110:3027-3037. [PMID: 31348591 PMCID: PMC6778629 DOI: 10.1111/cas.14144] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Revised: 07/22/2019] [Accepted: 07/22/2019] [Indexed: 12/16/2022] Open
Abstract
We previously established a method to generate myeloid cells with a proliferative capability from pluripotent stem cells and designated them iPS-ML. Human iPS-ML cells share features with physiological macrophages including the capability to infiltrate into cancer tissues. We observed therapeutic effects of human iPS-ML cells expressing interferon β (iPS-ML/interferon (IFN)-β) in xenograft cancer models. However, assessment of host immune system-mediated therapeutic and adverse effects of this therapy is impossible by xenograft models. We currently evaluated the therapeutic effects of a mouse equivalent of human iPS-ML/IFN, a mouse embryonic stem (ES) cell-derived myeloid cell line producing IFN (ES-ML/IFN). The ES-MLs producing IFN-β (β-ML) and IFN-γ (γ-ML) and originating from E14 ES cells derived from the 129 mouse strain (H-2b ) were generated, and the MHC (H-2Kb , Db , and I-Ab ) genes of the ES-ML/IFN were disrupted using the clustered regularly interspaced short palindromic repeats (CRISPR)/CAS9 method. We used the ES-ML/IFN to treat allogeneic BALB/c mice (H-2d ) transplanted with Colon26 cancer cells. Treatment with β-ML but not with γ-ML cells repressed the growth of colon cancer in the peritoneal cavity and liver. The transferred ES-ML/IFN infiltrated into cancer tissues and enhanced infiltration of T cells into cancer tissues. ES-ML/IFN therapy increased the number of immune cells in the lymphoid organs. Sensitization of both cancer antigen-specific CD8+ T cells and natural killer (NK) cells were enhanced by the therapy, and CD8+ T cells were essential for the therapeutic effect, implying that donor MHC-deficient β-ML exhibited a therapeutic effect through the activation of host immune cells derived from allogeneic recipient mice. The results suggested the usefulness of HLA-deficient human iPS-ML/IFN-β cells for therapy of HLA-mismatched allogeneic cancer patients.
Collapse
Affiliation(s)
- Satoshi Umemoto
- Department of Immunogenetics, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Miwa Haruta
- Department of Immunogenetics, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Masataka Sakisaka
- Department of Immunogenetics, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Tokunori Ikeda
- Department of Clinical Investigation, Kumamoto University Hospital, Kumamoto, Japan
| | - Hirotake Tsukamoto
- Department of Immunology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yoshihiro Komohara
- Department of Cell Pathology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Motohiro Takeya
- Department of Cell Pathology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yasuharu Nishimura
- Department of Immunogenetics, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan.,Nishimura Project Laboratory, Institute of Resource Development and Analysis, Kumamoto University, Kumamoto, Japan
| | - Satoru Senju
- Department of Immunogenetics, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
192
|
Abstract
Immunotherapy has dramatically improved the prognosis for patients with melanoma and has become the cornerstone of treatment for those with advanced disease. The role of immunotherapy continues to expand with multiple new agents approved in the adjuvant as well as metastatic setting, as first-line therapy and beyond. We review the currently approved drugs for the treatment of melanoma, along with clinical trial data, adverse side effects, response assessment and future directions.
Collapse
Affiliation(s)
- Emily Feld
- UDepartment of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Tara C Mitchell
- UDepartment of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
193
|
Khunger A, Buchwald ZS, Lowe M, Khan MK, Delman KA, Tarhini AA. Neoadjuvant therapy of locally/regionally advanced melanoma. Ther Adv Med Oncol 2019; 11:1758835919866959. [PMID: 31391869 PMCID: PMC6669845 DOI: 10.1177/1758835919866959] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 07/08/2019] [Indexed: 11/18/2022] Open
Abstract
Locally/regionally advanced melanoma confers a major challenge in terms of surgical and medical management. Surgical treatment carries the risks of surgical morbidities and potential complications that could be lasting. In addition, these patients continue to have a high risk of relapse and death despite the use of standard adjuvant therapy. Neoadjuvant therapy has the potential to significantly improve the clinical outcome of these patients, particularly in this era of newer and effective targeted and immunotherapeutic agents. Previous neoadjuvant studies tested chemotherapy with temozolomide where the clinical activity was limited. Biochemotherapy (BCT) was tested in two studies in the neoadjuvant setting and showed high tumor response rates; however, BCT was ultimately abandoned following its failure to demonstrate survival benefits in randomized trials of metastatic disease. Success of immunotherapy and targeted therapy in prolonging the lives of patients with metastatic melanoma generated considerable interest to investigate these novel strategies in the adjuvant and neoadjuvant settings. A number of neoadjuvant targeted and immunotherapy studies have been completed in melanoma to date and have yielded promising clinical activity. Given these encouraging results, a number of studies with other molecularly targeted and immunotherapeutic agents and their combinations are ongoing in the neoadjuvant setting; long-term outcome data are eagerly awaited. Such studies also provide access to biospecimens before and during therapy, allowing for the conduct of biomarker and mechanistic studies that may have a significant impact in guiding adjuvant therapy choices and drug development.
Collapse
Affiliation(s)
- Arjun Khunger
- Department of Hematology and Oncology, Cleveland Clinic Taussig Cancer Center, Cleveland, OH, USA
| | - Zachary S. Buchwald
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA, USA
| | - Michael Lowe
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, USA
| | - Mohammad K. Khan
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA, USA
| | - Keith A. Delman
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, USA
| | - Ahmad A. Tarhini
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Winship Comprehensive Cancer Center, 1365 Clifton Rd Atlanta, GA 30322, USA
| |
Collapse
|
194
|
Lorenzi M, Arndorfer S, Aguiar-Ibañez R, Scherrer E, Liu FX, Krepler C. An indirect treatment comparison of the efficacy of pembrolizumab versus competing regimens for the adjuvant treatment of stage III melanoma. J Drug Assess 2019; 8:135-145. [PMID: 31489255 PMCID: PMC6713115 DOI: 10.1080/21556660.2019.1649266] [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] [Received: 03/22/2019] [Accepted: 07/10/2019] [Indexed: 01/04/2023] Open
Abstract
Objective: To determine the efficacy of pembrolizumab relative to other treatments used in stage III melanoma by conducting a systematic literature review (SLR) and network meta-analysis (NMA). Methods: A SLR was conducted to identify randomized clinical trials (RCTs) evaluating approved adjuvant treatments including interferon-containing regimens, BRAF-inhibitors, and PD-L1 inhibitors in stage III melanoma patients. Relative treatment effects for recurrence-free survival (RFS) were synthesized with Bayesian NMA models that allowed for hazard ratios (HRs) to vary over time. Results: Included studies formed a connected network of evidence composed of eight trials. In high-risk stage III patients, the HR for pembrolizumab vs observation decreased significantly over time with the superiority of pembrolizumab over observation becoming statistically meaningful before 3 months. By 9 months, the HR for pembrolizumab vs observation was statistically significantly lower than the HR for most other treatments vs observation, with the exception of ipilimumab and biochemotherapy due to overlapping 95% credible intervals. In BRAF + patients, pembrolizumab was statistically significantly better than observation after 3 months. The HR for both BRAF-inhibitors vs observation increased significantly over time and pembrolizumab was statistically superior to both BRAF-inhibitors after 15 months. Conclusions: Pembrolizumab results in statistically significantly improved RFS compared to all competing regimens after 9 months, except ipilimumab and biochemotherapy, for the adjuvant treatment of stage III melanoma. However, point estimate HRs vs observation for pembrolizumab are much lower than those for ipilimumab. In BRAF + patients, the advantage of pembrolizumab versus competing interventions increases over time with respect to RFS.
Collapse
|
195
|
Abstract
The incidence of metastatic melanoma continues to increase each decade. Although surgical treatment is often curative for localized stage I and stage II disease, the median survival for patients with distant metastases is less than 1 year. The last 2 decades have witnessed a breakthrough in therapeutic options with the development of immune checkpoint inhibitors, small molecule targeted therapy, and oncolytic viral therapy. This article provides an overview of the treatment options available for advanced melanoma, including chemotherapy, targeted therapy, immunotherapy, interleukin-2, and oncolytic viral agents.
Collapse
Affiliation(s)
- Leonora Bomar
- Department of Dermatology, Wake Forest School of Medicine, Winston-Salem, NC, USA.
| | - Aditi Senithilnathan
- Department of Dermatology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Christine Ahn
- Department of Dermatology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
196
|
Güney İB, Küçüker KA. Cilt kanserlerinde lenfosintigrafi ve cerrahi gama prob ile sentinel lenf nodu biyopsisinin minimal invaziv cerrahi yaklaşımındaki rolü. CUKUROVA MEDICAL JOURNAL 2019. [DOI: 10.17826/cumj.411652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
|
197
|
Abstract
OPINION STATEMENT In recent years, the number of patients with malignant melanoma has continued to increase globally; surgery remains the first treatment option for patients with resectable melanoma. Adjuvant therapy for patients with stage III and IV melanoma following surgical resection has gradually been approved. After complete resection, these patients can probably derive significant benefit from adjuvant therapy. New treatments that improve the long-term survival of patients with unresectable advanced or metastatic melanoma are currently under evaluation in adjuvant therapy to increase relapse-free survival and overall survival. We here review several relevant clinical trials of radiotherapy, systemic immune therapies, molecular-targeted therapies, and neoadjuvant therapies in order to shed light on most suitable adjuvant therapy. The findings of this review include the following: The use of interferon-α2b will be restricted for patients with ulcerated primary melanoma in countries with no access to new drugs in adjuvant therapy. Ipilimumab should not be considered as the first-line therapy due to its lower efficacy and severe toxicity. The use of anti-programmed death-1 antibody would be a relevant adjuvant therapy for patients without BRAF mutation. If the BRAF mutation status is positive, the combination of dabrafenib and trametinib is a plausible option. The establishment of appropriate therapeutic planning and clinical endpoints in adjuvant therapy should affect the standard of care. The choice of optimal adjuvant therapy for individual patients is an important issue.
Collapse
Affiliation(s)
- Maiko Wada-Ohno
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, -1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan.
| | - Takamichi Ito
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, -1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Masutaka Furue
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, -1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| |
Collapse
|
198
|
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
|
199
|
Abstract
Interferon gamma has long been studied as a critical mediator of tumor immunity. In recent years, the complexity of cellular interactions that take place in the tumor microenvironment has become better appreciated in the context of immunotherapy. While checkpoint inhibitors have dramatically improved remission rates in cancer treatment, IFN-γ and related effectors continue to be identified as strong predictors of treatment success. In this review, we provide an overview of the multiple immunosuppressive barriers that IFN-γ has to overcome to eliminate tumors, and potential avenues for modulating the immune response in favor of tumor rejection.
Collapse
Affiliation(s)
- J Daniel Burke
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, USA.
| | - Howard A Young
- Laboratory of Experimental Immunology, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA
| |
Collapse
|
200
|
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
|