701
|
Murciano-Goroff YR, Taylor BS, Hyman DM, Schram AM. Toward a More Precise Future for Oncology. Cancer Cell 2020; 37:431-442. [PMID: 32289268 PMCID: PMC7499397 DOI: 10.1016/j.ccell.2020.03.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 03/12/2020] [Accepted: 03/16/2020] [Indexed: 12/11/2022]
Abstract
Prospective molecular characterization of cancer has enabled physicians to define the genomic changes of each patient's tumor in real time and select personalized therapies based on these detailed portraits. Despite the promise of such an approach, previously unrecognized biological and therapeutic complexity is emerging. Here, we synthesize lessons learned and discuss the steps required to extend the benefits of genome-driven oncology, including proposing strategies for improved drug design, more nuanced patient selection, and optimized use of available therapies. Finally, we suggest ways that next-generation genome-driven clinical trials can evolve to accelerate our understanding of cancer biology and improve patient outcomes.
Collapse
Affiliation(s)
- Yonina R Murciano-Goroff
- Department of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Barry S Taylor
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Human Oncogenesis and Pathology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Weill Cornell Medical College, New York, NY 10065, USA
| | - David M Hyman
- Department of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA; Weill Cornell Medical College, New York, NY 10065, USA; Loxo Oncology, A Wholly Owned Subsidiary of Eli Lilly, Stamford, CT, USA
| | - Alison M Schram
- Department of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA; Weill Cornell Medical College, New York, NY 10065, USA.
| |
Collapse
|
702
|
Jenkins RW, Fisher DE. Treatment of Advanced Melanoma in 2020 and Beyond. J Invest Dermatol 2020; 141:23-31. [PMID: 32268150 DOI: 10.1016/j.jid.2020.03.943] [Citation(s) in RCA: 213] [Impact Index Per Article: 42.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 03/16/2020] [Indexed: 01/22/2023]
Abstract
The melanoma field has seen an unprecedented set of clinical advances over the past decade. Therapeutic efficacy for advanced or metastatic melanoma went from being one of the most poorly responsive to one of the more responsive. Perhaps most strikingly, the advances that transformed management of the disease are based upon modern mechanism-based therapeutic strategies. The targeted approaches that primarily suppress the BRAF oncoprotein pathway have a high predictability of efficacy although less optimal depth or durability of response. Immunotherapy is primarily based on blockade of one or two immune checkpoints and has a lower predictability of response but higher fractions of durable remissions. This article reviews the clinical progress in management of advanced melanoma and also discusses the impact of the same therapies on earlier stage disease, where the agents have shown significant promise in treating resectable but high-risk clinical scenarios. Collectively, the progress in melanoma therapeutics has transformed the standard of care for patients, informed new approaches that are increasingly utilized for treatment of other malignancies, and suggest novel strategies to further boost efficacy for the many patients not yet receiving optimal benefit from these approaches.
Collapse
Affiliation(s)
- Russell W Jenkins
- Center for Cancer Research, Department of Medicine, MGH Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA; Laboratory for Systems Pharmacology, Harvard Program in Therapeutic Sciences, Harvard Medical School, Boston, Massachusetts, USA
| | - David E Fisher
- Cutaneous Biology Research Center, Department of Dermatology and MGH Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
703
|
Chorti E, Kanaki T, Zimmer L, Hadaschik E, Ugurel S, Gratsias E, Roesch A, Bonella F, Wessendorf TE, Wälscher J, Theegarten D, Schadendorf D, Livingstone E. Drug-induced sarcoidosis-like reaction in adjuvant immunotherapy: Increased rate and mimicker of metastasis. Eur J Cancer 2020; 131:18-26. [PMID: 32248071 DOI: 10.1016/j.ejca.2020.02.024] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 02/07/2020] [Accepted: 02/17/2020] [Indexed: 01/06/2023]
Abstract
BACKGROUND Anti-[programmed cell death protein 1 (PD-1)] antibodies nivolumab and pembrolizumab were approved for adjuvant treatment of melanoma as they demonstrated improved relapse-free survival. Currently, combined anti-PD-1 plus anti-[cytotoxic T-lymphocyte-associated protein 4 (CTLA4)] blockade is being investigated in adjuvant and neoadjuvant trials. Sarcoidosis-like reactions have been described for immune checkpoint inhibitors and are most likely drug-induced. The reported rate of sarcoidosis/sarcoidosis-like reactions within clinical melanoma trials is <2%. We observed that a remarkably higher number of melanoma patients (10/45 patients, 22%) treated with immune checkpoint inhibitor (ICI) within an adjuvant clinical trial-developed drug induced sarcoidosis-like reaction (DISR) mimicking metastasis. CASE PRESENTATION Of 45 stage III melanoma patients who were treated at our institute with adjuvant ICI (either nivolumab alone or in combination with ipilimumab) within a two-armed, blinded clinical trial, ten developed a DISR. Three of the ten patients were men, median age was 52 years (range, 32-70 years). DISRs were asymptomatic and generally detected radiographically at first radiographic imaging after the start of therapy (median time, 2.8 months) and described as a differential diagnosis to tumour progression. In one patient, DISR was only apparent 13.1 months after start of therapy and 4 weeks after the end of ICI treatment. DISR presented as mediastinal/hilar lymphadenopathy in 8/10 patients (as only site or in addition to lung, skin and/or bone involvement), one patient had only lung and cutaneous, one patient only cutaneous DISR. Biopsies from lymph nodes, skin and bone were taken in 8/10 patients, and histology confirmed sarcoidosis-like reactions (SLRs). As patients were asymptomatic, no treatment for DISR was required, and study treatment was stopped for DISR in only one patient due to bone involvement. DISRs have resolved or are in remission in all patients. At a median follow-up time of 15.3 months (range, 12-17.6 months), two patients experienced melanoma relapse. CONCLUSIONS In most cases, sarcoidosis could only be differentiated from melanoma progression on biopsy. Treating physicians as well as radiologists have to be aware of the potentially higher rate of DISR in patients receiving adjuvant ICI. A thorough interdisciplinary workup is required to discriminate from true melanoma progression and to decide on continuation of adjuvant ICI treatment.
Collapse
Affiliation(s)
- Eleftheria Chorti
- Dept. of Dermatology, University Hospital Essen, Hufelandstrasse 55, 45147 Essen, Germany.
| | - Theodora Kanaki
- Dept. of Dermatology, University Hospital Essen, Hufelandstrasse 55, 45147 Essen, Germany
| | - Lisa Zimmer
- Dept. of Dermatology, University Hospital Essen, Hufelandstrasse 55, 45147 Essen, Germany
| | - Eva Hadaschik
- Dept. of Dermatology, University Hospital Essen, Hufelandstrasse 55, 45147 Essen, Germany
| | - Selma Ugurel
- Dept. of Dermatology, University Hospital Essen, Hufelandstrasse 55, 45147 Essen, Germany
| | - Emmanouil Gratsias
- Dept. of Dermatology, University Hospital Essen, Hufelandstrasse 55, 45147 Essen, Germany
| | - Alexander Roesch
- Dept. of Dermatology, University Hospital Essen, Hufelandstrasse 55, 45147 Essen, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Francesco Bonella
- Dept. of Pulmonary Medicine, Center for Interstitial and Rare Lung Diseases, Ruhrlandklinik University Hospital Essen, Tüschener Weg 40, 45239 Essen, Germany
| | - Thomas E Wessendorf
- Dept. of Pulmonary Medicine, Center for Interstitial and Rare Lung Diseases, Ruhrlandklinik University Hospital Essen, Tüschener Weg 40, 45239 Essen, Germany
| | - Julia Wälscher
- Dept. of Pulmonary Medicine, Center for Interstitial and Rare Lung Diseases, Ruhrlandklinik University Hospital Essen, Tüschener Weg 40, 45239 Essen, Germany
| | - Dirk Theegarten
- Institute of Pathology, University Hospital Essen, Hufelandstrasse 55, 45147 Essen, Germany
| | - Dirk Schadendorf
- Dept. of Dermatology, University Hospital Essen, Hufelandstrasse 55, 45147 Essen, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Elisabeth Livingstone
- Dept. of Dermatology, University Hospital Essen, Hufelandstrasse 55, 45147 Essen, Germany
| |
Collapse
|
704
|
Nassar KW, Tan AC. The mutational landscape of mucosal melanoma. Semin Cancer Biol 2020; 61:139-148. [PMID: 31655118 PMCID: PMC7078020 DOI: 10.1016/j.semcancer.2019.09.013] [Citation(s) in RCA: 122] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 09/13/2019] [Accepted: 09/19/2019] [Indexed: 12/27/2022]
Abstract
Mucosal melanoma is a rare and aggressive subtype of melanoma that has a less favorable prognosis due to the lack of understanding and identification of oncogenic drivers. Recently, whole genome and whole exome sequencing have unveiled the molecular landscape and potential oncogenic drivers of mucosal melanoma, which remains distinct from cutaneous melanoma. In this review, we provide an overview of the genomic landscape of mucosal melanoma, with a focus on molecular studies identifying potential oncogenic drivers allowing for a better mechanistic understanding of the biology of mucosal melanoma. We summarized the published genomics and clinical data supporting the observations that mucosal melanoma harbors distinct genetic alterations and oncogenic drivers from cutaneous melanoma, and thus should be treated accordingly. The common drivers (BRAF and NRAS) found in cutaneous melanoma have lower mutation rate in mucosal melanoma. In contrast, SF3B1 and KIT have higher mutation rate in mucosal melanoma as compared to cutaneous melanoma. From the meta-analysis, we also observed that the mutational profiles are slightly different between the "upper" and "lower" regions of mucosal melanoma, providing new insights and therapeutic options for the mucosal melanoma patients. Mutations identified in mucosal melanoma should be incorporated into routine clinical testing, as there are targeted therapies already developed for treating patients with these mutations in the precision medicine era.
Collapse
Affiliation(s)
- Kelsey W Nassar
- Division of Medical Oncology, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80111, USA; Cancer Biology Training Program, Graduate School, University of Colorado Anschutz Medical Campus, Aurora, CO 80111, USA
| | - Aik Choon Tan
- Division of Medical Oncology, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80111, USA; Cancer Biology Training Program, Graduate School, University of Colorado Anschutz Medical Campus, Aurora, CO 80111, USA; Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, FL 33612 USA.
| |
Collapse
|
705
|
Totzeck M, Rassaf T. Neue onkologische Therapien und ihre kardiovaskulären Risiken. Herz 2020; 45:129-133. [DOI: 10.1007/s00059-020-04902-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
706
|
Moreno-Ramírez D, Vieira R, Kaufmann R. Surgical approach to patients with low-burden stage III melanoma: Is it time to consider conservative surgery? Eur J Surg Oncol 2020; 46:498-500. [DOI: 10.1016/j.ejso.2019.11.500] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 11/14/2019] [Indexed: 11/28/2022] Open
|
707
|
Yilmaz M, Güven Meşe Ş. Treatment exceeds expectations with vemurafenib monotherapy in a patient with BRAF V600E-mutant metastatic melanoma. J Oncol Pharm Pract 2020; 26:1754-1758. [PMID: 32223367 DOI: 10.1177/1078155220906011] [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: 11/15/2022]
Abstract
INTRODUCTION Patients with distant metastatic melanoma has a poor prognosis, with a reported median survival time of six to eight months. In modern era, survival has prolonged with the immunotherapy and targeted therapy options. Potent and selective BRAF inhibitors have been developed that specifically inhibit mutated BRAF over other RAF kinases. Vemurafenib was the first selective tyrosine kinase inhibitor developed to target the V600E allele of BRAF-mutant melanoma. CASE REPORT In this report, we present a case of BRAFV600E-mutant metastatic melanoma, which is being treated with vemurafenib monotherapy with complete response for about seven years. MANAGEMENT AND OUTCOME The patient is still being treated with vemurafenib and radiologic complete response is ongoing for about seven years. DISCUSSION Patients treated with BRAF inhibitors monotherapy had promising response rates and improvement in the progression-free survival and overall survival, but melanoma cells became resistant very quickly, affecting the progression. In this case, we present a case that has permanent response to vemurafenib monotherapy.
Collapse
Affiliation(s)
- Mesut Yilmaz
- Medical Oncology Department, Bakırköy Sadi Konuk Training and Research Hospital, İstanbul, Turkey
| | - Şermin Güven Meşe
- Medical Oncology Department, Yeditepe University Koşuyolu Hospital, İstanbul, Turkey
| |
Collapse
|
708
|
Madu MF, Franke V, Van de Wiel BA, Klop WMC, Jóźwiak K, van Houdt WJ, Wouters MWJM, van Akkooi ACJ. External validation of the American Joint Committee on Cancer 8th edition melanoma staging system: who needs adjuvant treatment? Melanoma Res 2020; 30:185-192. [PMID: 31651715 DOI: 10.1097/cmr.0000000000000643] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Now effective adjuvant therapy has arrived in melanoma, accurate staging and patient selection to optimize its risk/benefit ratio is crucial. The American Joint Committee on Cancer staging system is the most widely used and validated melanoma staging system, which recently released its 8th edition. We aimed to externally validate the prognostic and discriminatory ability for survival of the 8th edition compared to the 7th edition and evaluate prognostic factors. Prospective database of stage III melanoma (2000-2016). Prognostic factors for melanoma-specific survival and distant metastasis-free survival were analyzed. Survival differentiation of the 7th and 8th edition was assessed with log-rank tests and Cox proportional hazards models. Discriminatory ability was compared using the receiver operating characteristic and Akaike's Information Criterion. Six hundred forty patients were included (median follow-up 59 months). Median melanoma-specific survival was 138 months, distant metastasis-free survival 96 months. Age, Breslow thickness, ulceration of the primary tumor and number of positive lymph nodes (N) were independent prognostic parameters for distant metastasis-free survival and melanoma-specific survival. The 8th edition performed slightly better than the 7th edition in terms of survival discrimination but showed slightly worse distant metastasis-free survival and melanoma-specific survival differentiation between stage IIIA and IIIB. Sentinel node (SN) metastasis size cutoff of 1 mm differentiated survival in both 7th and 8th edition stage IIIA, showing excellent distant metastasis-free survival and melanoma-specific survival for patients with a SN metastasis size <1 mm. The 8th edition performed at least comparably, if not better than the 7th in terms of survival discrimination. However, survival in both 7th and 8th edition stage IIIA melanoma remains heterogeneous. EORTC SN tumor burden criteria can further stratify survival and help patient selection for adjuvant therapy.
Collapse
Affiliation(s)
| | | | | | | | - Katarzyna Jóźwiak
- Epidemiology and Biostatistics, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, The Netherlands
| | | | | | | |
Collapse
|
709
|
Robinson AV, Keeble C, Lo MCI, Thornton O, Peach H, Moncrieff MDS, Dewar DJ, Wade RG. The neutrophil-lymphocyte ratio and locoregional melanoma: a multicentre cohort study. Cancer Immunol Immunother 2020; 69:559-568. [PMID: 31974724 PMCID: PMC7113207 DOI: 10.1007/s00262-019-02478-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 12/31/2019] [Indexed: 01/04/2023]
Abstract
OBJECTIVES The neutrophil-lymphocyte ratio (NLR) is an inflammatory biomarker which is useful in cancer prognostication. We aimed to investigate the differences in baseline NLR between patients with localised and metastatic cutaneous melanoma and how this biomarker changed over time with the recurrence of disease. METHODS This multicentre cohort study describes patients treated for Stage I-III cutaneous melanoma over 10 years. The baseline NLR was measured immediately prior to surgery and again at the time of discharge or disease recurrence. The odds ratios (OR) for sentinel node involvement are estimated using mixed-effects logistic regression. The risk of recurrence is estimated using multivariable Cox regression. RESULTS Overall 1489 individuals were included. The mean baseline NLR was higher in patients with palpable nodal disease compared to those with microscopic nodal or localised disease (2.8 versus 2.4 and 2.3, respectively; p < 0.001). A baseline NLR ≥ 2.3 was associated with 30% higher odds of microscopic metastatic melanoma in the sentinel lymph node [adjusted OR 1.3 (95% CI 1.3, 1.3)]. Following surgery, 253 patients (18.7%) developed recurrent melanoma during surveillance although there was no statistically significant association between the baseline NLR and the risk of recurrence [adjusted HR 0.9 (0.7, 1.1)]. CONCLUSION The NLR is associated with the volume of melanoma at presentation and may predict occult sentinel lymph metastases. Further prospective work is required to investigate how NLR may be modelled against other clinicopathological variables to predict outcomes and to understand the temporal changes in NLR following surgery for melanoma.
Collapse
Affiliation(s)
- Alyss V Robinson
- Leeds Institute for Medical Research, University of Leeds, Leeds, UK
| | - Claire Keeble
- Leeds Institute for Data Analytics, University of Leeds, Leeds, UK
| | - Michelle C I Lo
- Plastic and Reconstructive Surgery Department, Norfolk and Norwich University Hospital NHS Trust, Norwich, UK
| | - Owen Thornton
- Trinity College Dublin, The University of Dublin, Dublin, Ireland
| | - Howard Peach
- Department of Plastic and Reconstructive Surgery, Leeds General Infirmary, Leeds, UK
| | - Marc D S Moncrieff
- Plastic and Reconstructive Surgery Department, Norfolk and Norwich University Hospital NHS Trust, Norwich, UK
- Norwich Medical School, University of East Anglia, Norwich, UK
| | - Donald J Dewar
- Department of Plastic and Reconstructive Surgery, Leeds General Infirmary, Leeds, UK
| | - Ryckie G Wade
- Leeds Institute for Medical Research, University of Leeds, Leeds, UK.
- Department of Plastic and Reconstructive Surgery, Leeds General Infirmary, Leeds, UK.
| |
Collapse
|
710
|
Madorsky Rowdo FP, Barón A, Gallagher SJ, Hersey P, Emran AA, Von Euw EM, Barrio MM, Mordoh J. Epigenetic inhibitors eliminate senescent melanoma BRAFV600E cells that survive long‑term BRAF inhibition. Int J Oncol 2020; 56:1429-1441. [PMID: 32236593 PMCID: PMC7170042 DOI: 10.3892/ijo.2020.5031] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 02/05/2020] [Indexed: 02/06/2023] Open
Abstract
It is estimated that ~50% of patients with melanoma harbour B‑Raf (BRAF)V600 driver mutations, with the most common of these being BRAFV600E, which leads to the activation of mitogen‑activated protein kinase proliferative and survival pathways. BRAF inhibitors are used extensively to treat BRAF‑mutated metastatic melanoma; however, acquired resistance occurs in the majority of patients. The effects of long‑term treatment with PLX4032 (BRAFV600 inhibitor) were studied in vitro on sensitive V600E BRAF‑mutated melanoma cell lines. After several weeks of treatment with PLX4032, the majority of the melanoma cells died; however, a proportion of cells remained viable and quiescent, presenting senescent cancer stem cell‑like characteristics. This surviving population was termed SUR cells, as discontinuing treatment allowed the population to regrow while retaining equal drug sensitivity to that of parental cells. RNA sequencing analysis revealed that SUR cells exhibit changes in the expression of 1,415 genes (P<0.05) compared with parental cells. Changes in the expression levels of a number of epigenetic regulators were also observed. These changes and the reversible nature of the senescence state were consistent with epigenetic regulation; thus, it was investigated as to whether the senescent state could be reversed by epigenetic inhibitors. It was found that both parental and SUR cells were sensitive to different histone deacetylase (HDAC) inhibitors, such as SAHA and MGCD0103, and to the cyclin‑dependent kinase (CDK)9 inhibitor, CDKI‑73, which induced apoptosis and reduced proliferation both in the parental and SUR populations. The results suggested that the combination of PLX4032 with HDAC and CDK9 inhibitors may achieve complete elimination of SUR cells that persist after BRAF inhibitor treatment, and reduce the development of resistance to BRAF inhibitors.
Collapse
Affiliation(s)
- Florencia Paula Madorsky Rowdo
- Cancerology Laboratory, Leloir Institute‑Biochemical Research Institute of Buenos Aires (IIBBA), National Scientific and Technical Research Council (CONICET), Buenos Aires C1405BWE, Argentina
| | - Antonela Barón
- Cancerology Laboratory, Leloir Institute‑Biochemical Research Institute of Buenos Aires (IIBBA), National Scientific and Technical Research Council (CONICET), Buenos Aires C1405BWE, Argentina
| | - Stuart John Gallagher
- Melanoma Oncology and Immunology Group, Centenary Institute, Sydney, New South Wales 2050, Australia
| | - Peter Hersey
- Melanoma Oncology and Immunology Group, Centenary Institute, Sydney, New South Wales 2050, Australia
| | - Abdullah Al Emran
- Melanoma Oncology and Immunology Group, Centenary Institute, Sydney, New South Wales 2050, Australia
| | - Erika M Von Euw
- Department of Medicine, Division of Hematology‑Oncology, Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA 90024, USA
| | - María Marcela Barrio
- Oncology Research Center‑Cancer Foundation (FUCA), Buenos Aires C1426 ANZ, Argentina
| | - José Mordoh
- Cancerology Laboratory, Leloir Institute‑Biochemical Research Institute of Buenos Aires (IIBBA), National Scientific and Technical Research Council (CONICET), Buenos Aires C1405BWE, Argentina
| |
Collapse
|
711
|
Identification of Small Molecule Enhancers of Immunotherapy for Melanoma. Sci Rep 2020; 10:5688. [PMID: 32231230 PMCID: PMC7105471 DOI: 10.1038/s41598-020-62369-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 03/12/2020] [Indexed: 02/01/2023] Open
Abstract
Small molecule based targeted therapies for the treatment of metastatic melanoma hold promise but responses are often not durable, and tumors frequently relapse. Response to adoptive cell transfer (ACT)-based immunotherapy in melanoma patients are durable but patients develop resistance primarily due to loss of antigen expression. The combination of small molecules that sustain T cell effector function with ACT could lead to long lasting responses. Here, we have developed a novel co-culture cell-based high throughput assay system to identify compounds that could potentially synergize or enhance ACT-based immunotherapy of melanoma. A BRAFV600E mutant melanoma cell line, SB-3123p which is resistant to Pmel-1-directed ACT due to low gp100 expression levels was used to develop a homogenous time resolve fluorescence (HTRF), screening assay. This high throughput screening assay quantitates IFNγ released upon recognition of the SB-3123p melanoma cells by Pmel-1 CD8+ T-cells. A focused collection of approximately 500 small molecules targeting a broad range of cellular mechanisms was screened, and four active compounds that increased melanoma antigen expression leading to enhanced IFNγ production were identified and their in vitro activity was validated. These four compounds may provide a basis for enhanced immune recognition and design of novel therapeutic approaches for patients with BRAF mutant melanoma resistant to ACT due to antigen downregulation.
Collapse
|
712
|
van Willigen WW, Bloemendal M, Boers-Sonderen MJ, de Groot JWB, Koornstra RHT, van der Veldt AAM, Haanen JBAG, Boudewijns S, Schreibelt G, Gerritsen WR, de Vries IJM, Bol KF. Response and survival of metastatic melanoma patients treated with immune checkpoint inhibition for recurrent disease on adjuvant dendritic cell vaccination. Oncoimmunology 2020; 9:1738814. [PMID: 33457087 PMCID: PMC7790511 DOI: 10.1080/2162402x.2020.1738814] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Vaccination with autologous dendritic cells (DC) loaded ex vivo with melanoma-associated antigens is currently being tested as an adjuvant treatment modality for resected locoregional metastatic (stage III) melanoma. Based on its mechanism of action, DC vaccination might potentiate the clinical efficacy of concurrent or sequential immune checkpoint inhibition (ICI). The purpose of this study was to determine the efficacy of ICI administered following recurrent disease during, or after, adjuvant DC vaccination. To this end, we retrospectively analyzed clinical responses of 51 melanoma patients with either irresectable stage III or stage IV disease treated with first- or second-line ICI following recurrence on adjuvant DC vaccination. Patients were analyzed according to the form of ICI administered: PD-1 inhibition monotherapy (nivolumab or pembrolizumab), ipilimumab monotherapy or combined treatment with ipilimumab and nivolumab. Treatment with first- or second-line PD-1 inhibition monotherapy after recurrence on adjuvant DC vaccination resulted in a response rate of 52%. In patients treated with ipilimumab monotherapy and ipilimumab-nivolumab response rates were 35% and 75%, respectively. In conclusion, ICI is effective in melanoma patients with recurrent disease on adjuvant DC vaccination.
Collapse
Affiliation(s)
- Wouter W van Willigen
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, The Netherlands.,Department of Medical Oncology, Radboudumc, Nijmegen, The Netherlands
| | - Martine Bloemendal
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, The Netherlands.,Department of Medical Oncology, Radboudumc, Nijmegen, The Netherlands
| | | | | | - Rutger H T Koornstra
- Department of Medical Oncology, Radboudumc, Nijmegen, The Netherlands.,Department of Internal Medicine, Hospital Rijnstate, Arnhem, The Netherlands
| | - Astrid A M van der Veldt
- Department of Medical Oncology, Erasmus Medical Center Cancer Institute, Rotterdam, The Netherlands.,Department of Radiology & Nuclear Medicine, Erasmus Medical Center Cancer Institute, Rotterdam, The Netherlands
| | - John B A G Haanen
- Division of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Steve Boudewijns
- Department of Medical Oncology, Bravis Hospital, Roosendaal, The Netherlands
| | - Gerty Schreibelt
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, The Netherlands
| | | | - I Jolanda M de Vries
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, The Netherlands.,Department of Medical Oncology, Radboudumc, Nijmegen, The Netherlands
| | - Kalijn F Bol
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, The Netherlands.,Department of Medical Oncology, Radboudumc, Nijmegen, The Netherlands
| |
Collapse
|
713
|
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
|
714
|
Theile H, Moore J, Dunn N, Cossio D, Forristal CE, Green AC, Smithers BM. Regional nodal metastasis and 5-year survival in patients with thin melanoma in Queensland: a population-based study. ANZ J Surg 2020; 90:503-507. [PMID: 32162780 DOI: 10.1111/ans.15804] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 12/16/2019] [Accepted: 02/20/2020] [Indexed: 11/28/2022]
Abstract
BACKGROUND Optimal management of regional lymph nodes for thin cutaneous melanoma is uncertain. We evaluated regional lymph node involvement and 5-year melanoma-specific survival (MSS) in patients with thin (≤1 mm) primary melanoma. METHODS Patients with a melanoma, American Joint Committee on Cancer Staging 8th Edition pT1a (<0.8 mm) or pT1b (ulceration; and/or 0.8-1.0 mm), diagnosed during 2001-2015 were identified from the Queensland Oncology Repository. We extracted demographic, pathology and clinical details, including sentinel lymph node biopsy (SLNB), regional nodal dissection and nodal recurrence. Poisson regression was used to assess recurrence risk in patients who did not undergo SLNB. The 5-year MSS was calculated using the Kaplan-Maier method with Cox regression to compare survival outcomes according to SLNB performance. RESULTS Of the 27 824 eligible patients, 240 (0.9%) underwent SLNB. One hundred and seventy-eight patients (0.6%) without SLNB had nodal recurrence. Of the 4848 patients with a pT1b lesion, 166 (3.4%) had SLNB with 12 (7.2%) positive; of the remainder, 99 (2.1%) had clinical recurrence. Risk of recurrence was higher in males, nodular subtype and T1b lesions and lower if patients were aged >60 years. The 5-year MSS was similar for observed and SLNB cohorts (99.66% versus 98.92%) but worse for T1b lesions (98.90%) and clinical nodal recurrence (66.89%). CONCLUSION Overall prognosis for T1 melanoma is excellent with nodal involvement being rare. However, the American Joint Committee on Cancer 8th Edition T1b melanoma correlates with significantly worse 5-year MSS and increased regional nodal recurrence (notably for 0.8-1.0 mm lesions with ulceration). Further characterization of high-risk groups for nodal positivity that impacts patient outcome is needed for the pT1 melanoma cohort.
Collapse
Affiliation(s)
- Harrison Theile
- Discipline of Surgery, The University of Queensland, Brisbane, Queensland, Australia.,Queensland Cancer Control Analysis Team, Cancer Alliance Queensland, Brisbane, Queensland, Australia.,Queensland Melanoma Project, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Julie Moore
- Queensland Cancer Control Analysis Team, Cancer Alliance Queensland, Brisbane, Queensland, Australia
| | - Nathan Dunn
- Queensland Cancer Control Analysis Team, Cancer Alliance Queensland, Brisbane, Queensland, Australia
| | - Danica Cossio
- Queensland Cancer Control Analysis Team, Cancer Alliance Queensland, Brisbane, Queensland, Australia
| | - Catherine E Forristal
- Queensland Melanoma Collaborative, Brisbane, Queensland, Australia.,Mater Research Institute, Brisbane, Queensland, Australia
| | - Adele C Green
- Population Health Department, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia.,CRUK Manchester Institute, University of Manchester, Manchester, UK
| | - B Mark Smithers
- Discipline of Surgery, The University of Queensland, Brisbane, Queensland, Australia.,Queensland Melanoma Project, Princess Alexandra Hospital, Brisbane, Queensland, Australia.,Queensland Melanoma Collaborative, Brisbane, Queensland, Australia.,Mater Research Institute, Brisbane, Queensland, Australia
| |
Collapse
|
715
|
Urbani F, Ferraresi V, Capone I, Macchia I, Palermo B, Nuzzo C, Torsello A, Pezzotti P, Giannarelli D, Pozzi AF, Santaquilani M, Roazzi P, Bastucci S, Catricalà C, La Malfa A, Vercillo G, Gualtieri N, Buccione C, Castiello L, Cognetti F, Nisticò P, Belardelli F, Moschella F, Proietti E. Clinical and Immunological Outcomes in High-Risk Resected Melanoma Patients Receiving Peptide-Based Vaccination and Interferon Alpha, With or Without Dacarbazine Preconditioning: A Phase II Study. Front Oncol 2020; 10:202. [PMID: 32211314 PMCID: PMC7069350 DOI: 10.3389/fonc.2020.00202] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 02/06/2020] [Indexed: 12/12/2022] Open
Abstract
Clinical studies based on novel rationales and mechanisms of action of chemotherapy agents and cytokines can contribute to the development of new concepts and strategies of antitumor combination therapies. In previous studies, we investigated the paradoxical immunostimulating effects of some chemotherapeutics and the immunoadjuvant activity of interferon alpha (IFN-α) in preclinical and clinical models, thus unraveling novel rationales and mechanisms of action of chemotherapy agents and cytokines for cancer immunotherapy. Here, we carried out a randomized, phase II clinical trial, in which we analyzed the relapse-free (RFS) and overall survival (OS) of 34 completely resected stage III–IV melanoma patients, treated with peptide-based vaccination (Melan-A/MART-1 and NY-ESO-1) in combination with IFN-α2b, with (arm 2) or without (arm 1) dacarbazine preconditioning. All patients were included in the intention-to-treat analysis. At a median follow-up of 4.5 years (interquartile range, 15.4–81.0 months), the rates of RFS were 52.9 and 35.3% in arms 1 and 2, respectively. The 4.5-year OS rates were 68.8% in arm 1 and 62.7% in arm 2. No significant differences were observed between the two arms for both RFS and OS. Interestingly, the RFS and OS curves remained stable starting from 18 and 42 months, respectively. Grade 3 adverse events occurred in 5.9% of patients, whereas grade 4 events were not observed. Both treatments induced a significant expansion of vaccine-specific CD8+ T cells, with no correlation with the clinical outcome. However, treatment-induced increase of polyfunctionality and of interleukin 2 production by Melan-A–specific CD8+ T cells and expansion/activation of natural killer cells correlated with RFS, being observed only in nonrelapsing patients. Despite the recent availability of different therapeutic options, low-cost, low-toxic therapies with long-lasting clinical effects are still needed in patients with high-risk resected stage III/IV melanoma. The combination of peptide vaccination with IFN-α2b showed a minimal toxicity profile and resulted in encouraging RFS and OS rates, justifying further evaluation in clinical trials, which may include the use of checkpoint inhibitors to further expand the antitumor immune response and the clinical outcome. Clinical Trial Registration:https://www.clinicaltrialsregister.eu/ctr-search/search, identifier: 2008-008211-26
Collapse
Affiliation(s)
- Francesca Urbani
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy.,Medical Biotechnology and Translational Medicine, Tor Vergata University, Rome, Italy
| | - Virginia Ferraresi
- Department of Medical Oncology 1, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Imerio Capone
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Iole Macchia
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Belinda Palermo
- Unit of Tumor Immunology and Immunotherapy, Department of Research, Advanced Diagnostics and Technological Innovation, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Carmen Nuzzo
- Department of Medical Oncology 1, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Angela Torsello
- Department of Medical Oncology 1, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Patrizio Pezzotti
- Department of Infectious Disease, Istituto Superiore di Sanità, Rome, Italy
| | - Diana Giannarelli
- Biostatistical Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Anna Fausta Pozzi
- Hospital Pharmacia, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | | | - Paolo Roazzi
- Health Technology Assessement, Istituto Superiore di Sanità, Rome, Italy
| | - Silvia Bastucci
- Clinical Trial Center, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | | | - Antonia La Malfa
- Hospital Pharmacia, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Giuseppe Vercillo
- Clinical Pathology, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Novella Gualtieri
- Unit of Tumor Immunology and Immunotherapy, Department of Research, Advanced Diagnostics and Technological Innovation, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Carla Buccione
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | | | - Francesco Cognetti
- Department of Medical Oncology 1, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Paola Nisticò
- Unit of Tumor Immunology and Immunotherapy, Department of Research, Advanced Diagnostics and Technological Innovation, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | | | - Federica Moschella
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Enrico Proietti
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
716
|
Dimou A, Barron G, Merrick DT, Kolfenbach J, Doebele RC. Granulomatosis with polyangiitis in a patient treated with dabrafenib and trametinib for BRAF V600E positive lung adenocarcinoma. BMC Cancer 2020; 20:177. [PMID: 32131760 PMCID: PMC7057580 DOI: 10.1186/s12885-020-6661-6] [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: 10/12/2018] [Accepted: 02/20/2020] [Indexed: 12/27/2022] Open
Abstract
Background Dabrafenib and trametinib combination therapy is approved for the treatment of patients with BRAF V600E positive tumors including melanoma and lung cancer. The effect of BRAF and MEK inhibitors on the immune system is not fully understood although a number of case reports indicate autoimmune side effects related to the use of these drugs. Here, we discuss a case of a patient diagnosed with granulomatosis with polyangiitis (GPA) shortly after starting treatment with dabrafenib and trametinib for BRAF V600E positive metastatic lung adenocarcinoma. Case presentation A 57 years old female patient was diagnosed with recurrent lung adenocarcinoma following initial lobectomy for early stage disease. A BRAF V600E mutation was identified at the time of recurrence and she received combination dabrafenib and trametinib therapy. Shortly after commencement of treatment, she developed persistent fevers necessitating withholding both drugs. Pyrexia continued and was followed by left vision loss and acute kidney injury. Further rheumatological workup led to the unifying diagnosis of GPA. The patient was then treated with rituximab for GPA to the present date while all antineoplastic drugs were held. Lung cancer oligoprogression was addressed with radiation therapy and has not required further systemic treatment whereas GPA has been controlled to-date with rituximab. Conclusions This case report raises awareness among clinicians treating patients with lung cancer for the possibility of triggering a flare of autoimmune diseases like GPA in patients with BRAF V600E positive lung cancer receiving treatment with BRAF directed therapy.
Collapse
Affiliation(s)
- Anastasios Dimou
- Division of Medical Oncology, University of Colorado, Anschutz Medical Campus, 13001 E 17th Pl, Aurora, CO, 80045, USA.
| | - Gregory Barron
- Division of Rheumatology, University of Colorado, Anschutz Medical Campus, 13001 E 17th Pl, Aurora, CO, 80045, USA
| | - Daniel T Merrick
- Department of Pathology, University of Colorado, School of Medicine, 13001 E 17th Pl, Aurora, CO, 80045, USA
| | - Jason Kolfenbach
- Division of Rheumatology, University of Colorado, Anschutz Medical Campus, 13001 E 17th Pl, Aurora, CO, 80045, USA
| | - Robert C Doebele
- Thoracic Oncology Research Initiative, Division of Medical Oncology, University of Colorado, School of Medicine, 12801 E. 17th Ave., MS 8117, Aurora, CO, 80045, USA
| |
Collapse
|
717
|
Huijberts S, Wang L, de Oliveira RL, Rosing H, Nuijen B, Beijnen J, Bernards R, Schellens J, Wilgenhof S. Vorinostat in patients with resistant BRAFV600E mutated advanced melanoma: a proof of concept study. Future Oncol 2020; 16:619-629. [PMID: 32125175 DOI: 10.2217/fon-2020-0023] [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] [Indexed: 12/21/2022] Open
Abstract
The clinical benefit of treatment with BRAF- and MEK-inhibitors in melanoma is limited due to resistance associated with emerging secondary mutations. Preclinical and clinical studies have shown that short-term treatment with the HDAC inhibitor vorinostat can eliminate cells harboring these secondary mutations causing resistance. This proof of concept study is to determine the efficacy of sequential treatment with vorinostat and BRAFi/MEKi in resistant BRAFV600E mutant melanoma. The primary aim is demonstrating anti-tumor response of progressive lesions according to RECIST 1.1. Secondary end points are to determine that emerging resistant clones with a secondary mutation in the MAPK pathway can be detected in circulating tumor DNA and purged by short-term vorinostat treatment. Exploratory end points include pharmacokinetic, pharmacodynamic and pharmacogenetic analyses (NCT02836548).
Collapse
Affiliation(s)
- Sanne Huijberts
- Department of Clinical Pharmacology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, The Netherlands
| | - Liqin Wang
- Division of Molecular Carcinogenesis, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, The Netherlands
| | - Rodrigo Leite de Oliveira
- Division of Molecular Carcinogenesis, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, The Netherlands
| | - Hilde Rosing
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, The Netherlands
| | - Bastiaan Nuijen
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, The Netherlands
| | - Jos Beijnen
- Department of Clinical Pharmacology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, The Netherlands.,Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, The Netherlands.,Faculty of Science, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
| | - Rene Bernards
- Division of Molecular Carcinogenesis, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, The Netherlands.,Faculty of Science, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
| | - Jan Schellens
- Faculty of Science, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
| | - Sofie Wilgenhof
- Department of Medical Oncology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, The Netherlands
| |
Collapse
|
718
|
Garzón-Orjuela N, Prieto-Pinto L, Lasalvia P, Herrera D, Castrillón J, González-Bravo D, Castañeda-Cardona C, Rosselli D. Efficacy and safety of dabrafenib-trametinib in the treatment of unresectable advanced/metastatic melanoma with BRAF-V600 mutation: A systematic review and network meta-analysis. Dermatol Ther 2020; 33:e13145. [PMID: 31664762 DOI: 10.1111/dth.13145] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 10/18/2019] [Accepted: 10/26/2019] [Indexed: 12/19/2022]
Abstract
The current systematic review aimed to evaluate and compare the efficacy and safety of dabrafenib-trametinib with those of other therapeutic alternatives in the treatment of patients with unresectable advanced/metastatic melanoma with BRAF-V600 mutation. The search was carried out on four databases up to July 2018. Two separate network meta-analyses (NMA) were performed using the frequentist method (random effects): one with an exclusive population with BRAF-V600 mutation (NMA-pBRAFV600) and another with mixed population (with or without the mutation: NMA-pMixed). An evidence profile was included using the GRADE method for NMA. The validity of the final estimator in the NMA-pMixed was assessed via a sensitivity analysis. Nine clinical trials were included in the NMA-pBRAFV600. Dabrafenib-trametinib was found to have a favorable effect on overall survival (OS) and progression-free survival (PFS) compared with dabrafenib, vemurafenib, and dacarbazine and on partial response rate (PRR) and overall response rate compared with dacarbazine and vemurafenib. In the NMA-pMixed, dabrafenib-trametinib was found to have a positive effect on OS versus ipilimumab 3 mg/kg and on PFS and PRR versus ipilimumab, nivolumab, and pembrolizumab. However, dabrafenib-trametinib and vemurafenib-cobimetinib significantly differed in terms of efficacy. In addition, dabrafenib-trametinib has a favorable effect on Grades 3 and 4 adverse events.
Collapse
Affiliation(s)
| | | | | | - Daniel Herrera
- Department of Medical - Oncology Unit, Novartis, Bogotá, Colombia
| | | | | | | | - Diego Rosselli
- Department of Evidence-Based Medicine, NeuroEconomix, Bogotá, Colombia
- Clinical Epidemiology and Biostatistics Department, Pontificia Universidad Javeriana, Medical School, Bogotá, Colombia
| |
Collapse
|
719
|
Schreck KC, Patel MP, Wemmer J, Grossman SA, Peters KB. RAF and MEK inhibitor therapy in adult patients with brain tumors: a case-based overview and practical management of adverse events. Neurooncol Pract 2020; 7:369-375. [PMID: 32765888 DOI: 10.1093/nop/npaa006] [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] [Indexed: 12/12/2022] Open
Abstract
Targeted therapy has gained mainstream attention with notable successes against specific genetic mutations in many cancers. One particular mutation, the BRAF V600E mutation, is present in a small subset of gliomas in adults. Although clinical experience and trial data of RAF-targeted therapy in adults with glioma are lacking at this time, the poor prognosis of adult high-grade glioma has led neuro-oncology practitioners to consider the use of targeted therapy in these patients. In this manuscript, we describe the use of RAF and MEK inhibitors in adults with recurrent glioma. We discuss the utility of these agents, describe their toxicities, and give examples of management strategies. Given the significant toxicities of RAF and MEK inhibitors, along with the long potential duration of treatment, neuro-oncology providers should counsel patients carefully before initiating therapy and monitor them closely while undergoing treatment with RAF-targeted therapy.
Collapse
Affiliation(s)
- Karisa C Schreck
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland, USA
| | - Mallika P Patel
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina, USA
| | - Jan Wemmer
- Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland, USA
| | | | - Katherine B Peters
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|
720
|
Rapport F, Smith AL, Cust AE, Mann GJ, Watts CG, Gyorki DE, Henderson M, Hong AM, Kelly JW, Long GV, Mar VJ, Morton RL, Saw RP, Scolyer RA, Spillane AJ, Thompson JF, Braithwaite J. Identifying challenges to implementation of clinical practice guidelines for sentinel lymph node biopsy in patients with melanoma in Australia: protocol paper for a mixed methods study. BMJ Open 2020; 10:e032636. [PMID: 32111612 PMCID: PMC7050375 DOI: 10.1136/bmjopen-2019-032636] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 12/24/2019] [Accepted: 01/28/2020] [Indexed: 11/04/2022] Open
Abstract
INTRODUCTION Sentinel lymph node biopsy (SLNB) is a diagnostic procedure developed in the 1990s. It is currently used to stage patients with primary cutaneous melanoma, provide prognostic information and guide management. The Australian Clinical Practice Guidelines state that SLNB should be considered for patients with cutaneous melanoma >1 mm in thickness (or >0.8 mm with high-risk pathology features). Until recently, sentinel lymph node (SLN) status was used to identify patients who might benefit from a completion lymph node dissection, a procedure that is no longer routinely recommended. SLN status is now also being used to identify patients who might benefit from systemic adjuvant therapies such as anti-programmed cell death 1 (PD1) checkpoint inhibitor immunotherapy or BRAF-directed molecular targeted therapy, treatments that have significantly improved relapse-free survival for patients with resected stage III melanoma and improved overall survival of patients with unresectable stage III and stage IV melanoma. Australian and international data indicate that approximately half of eligible patients receive an SLNB. METHODS AND ANALYSIS This mixed-methods study seeks to understand the structural, contextual and cultural factors affecting implementation of the SLNB guidelines. Data collection will include: (1) cross-sectional questionnaires and semistructured interviews with general practitioners and dermatologists; (2) semistructured interviews with other healthcare professionals involved in the diagnosis and early definitive care of melanoma patients and key stakeholders including researchers, representatives of professional colleges, training organisations and consumer melanoma groups; and (3) documentary analysis of documents from government, health services and non-government organisations. Descriptive analyses and multivariable regression models will be used to examine factors related to SLNB practices and attitudes. Qualitative data will be analysed using thematic analysis. ETHICS AND DISSEMINATION Ethics approval has been granted by the University of Sydney. Results will be disseminated through publications and presentations to clinicians, patients, policymakers and researchers and will inform the development of strategies for implementing SLNB guidelines in Australia.
Collapse
Affiliation(s)
- Frances Rapport
- Australian Institute of Health Innovation, Macquarie University, Sydney, New South Wales, Australia
| | - Andrea L Smith
- Australian Institute of Health Innovation, Macquarie University, Sydney, New South Wales, Australia
- Sydney School of Public Health, University of Sydney, Sydney, New South Wales, Australia
| | - Anne E Cust
- Sydney School of Public Health, University of Sydney, Sydney, New South Wales, Australia
- Melanoma Institute Australia, Sydney, New South Wales, Australia
| | - Graham J Mann
- Melanoma Institute Australia, Sydney, New South Wales, Australia
- The John Curtin School of Medical Research, Australian National Univeristy, Canberra, Australian Capital Territory, Australia
| | - Caroline G Watts
- Sydney School of Public Health, University of Sydney, Sydney, New South Wales, Australia
- Kirby Institute, University of New South Wales, Sydney, New South Wales, Australia
| | - David E Gyorki
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | | | - Angela M Hong
- Melanoma Institute Australia, Sydney, New South Wales, Australia
- Sydney School of Medicine, University of Sydney, Sydney, New South Wales, Australia
| | - John W Kelly
- Victorian Melanoma Service, The Alfred Hosptial, Melbourne, Victoria, Australia
| | - Georgina V Long
- Melanoma Institute Australia, Sydney, New South Wales, Australia
- Sydney School of Medicine, University of Sydney, Sydney, New South Wales, Australia
| | - Victoria J Mar
- Victorian Melanoma Service, The Alfred Hosptial, Melbourne, Victoria, Australia
| | - Rachael L Morton
- Melanoma Institute Australia, Sydney, New South Wales, Australia
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, New South Wales, Australia
| | - Robyn Pm Saw
- Melanoma Institute Australia, Sydney, New South Wales, Australia
- Sydney School of Medicine, University of Sydney, Sydney, New South Wales, Australia
| | - Richard A Scolyer
- Melanoma Institute Australia, Sydney, New South Wales, Australia
- Sydney School of Medicine, University of Sydney, Sydney, New South Wales, Australia
| | - Andrew J Spillane
- Melanoma Institute Australia, Sydney, New South Wales, Australia
- Sydney School of Medicine, University of Sydney, Sydney, New South Wales, Australia
| | - John F Thompson
- Melanoma Institute Australia, Sydney, New South Wales, Australia
- Sydney School of Medicine, University of Sydney, Sydney, New South Wales, Australia
| | - Jeffrey Braithwaite
- Australian Institute of Health Innovation, Macquarie University, Sydney, New South Wales, Australia
| |
Collapse
|
721
|
Krayem M, Aftimos P, Najem A, van den Hooven T, van den Berg A, Hovestad-Bijl L, de Wijn R, Hilhorst R, Ruijtenbeek R, Sabbah M, Kerger J, Awada A, Journe F, Ghanem GE. Kinome Profiling to Predict Sensitivity to MAPK Inhibition in Melanoma and to Provide New Insights into Intrinsic and Acquired Mechanism of Resistance. Cancers (Basel) 2020; 12:E512. [PMID: 32098410 PMCID: PMC7072684 DOI: 10.3390/cancers12020512] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 02/19/2020] [Accepted: 02/20/2020] [Indexed: 12/13/2022] Open
Abstract
Mitogen-activated protein kinase (MAPK) inhibition with the combination of BRAF (Rapidly Accelerated Fibrosarcoma) and MEK (Mitogen-activated protein kinase kinase) inhibitors has become the standard of first-line therapy of metastatic melanoma harbouring BRAF V600 mutations. However, about half of the patients present with primary resistance while the remaining develop secondary resistance under prolonged treatment. Thus, there is a need for predictive biomarkers for sensitivity and/or resistance to further refine the patient population likely to benefit from MAPK inhibitors. In this study, we explored a top-down approach using a multiplex kinase assay, first, to discover a kinome signature predicting sensitivity, intrinsic and acquired resistance to MAPK inhibitors in melanoma, and second, to understand the mechanism of resistance using cell lines. Pre-dose tissues from patients (four responders and three non-responders to BRAFi monotherapy) were profiled for phosphotyrosine kinase (PTK) and serine-threonine kinase (STK) activities on a PamChip® peptide microarray in the presence and absence of ex vivo BRAFi. In addition, molecular studies were conducted on four sensitive parental lines, their offspring with acquired resistance to BRAFi and two lines with intrinsic resistance. PTK and STK activities in cell lysates were measured in the presence and absence of ex vivo BRAFi and/or MEKi. In tissue lysates, concentration-dependent ex vivo inhibition of STK and PTK activities with dabrafenib was stronger in responders than in non-responders. This difference was confirmed in cell lines comparing sensitive and resistant ones. Interestingly, common features of resistance were increased activity of receptor tyrosine kinases, Proto-oncogene tyrosine-protein kinase Src (Src) family kinases and protein kinase B (PKB, AKT) signalling. These latter results were confirmed by Western blots. While dabrafenib alone showed an inhibition of STK and PTK activities in both tissues and cell lines, the combination of dabrafenib and trametinib showed an antagonism on the STK activities and a synergism on PTK activities, resulting in stronger inhibitions of overall tyrosine kinase activities. Altogether; these data reveal that resistance of tumours and cell lines to MAPK inhibitors can be predicted using a multiplex kinase assay and is associated with an increase in specific tyrosine kinase activities and globally to AKT signalling in the patient's tissue. Thus, such a predictive kinome signature would help to identify patients with innate resistance to MAPK double inhibition in order to propose other therapies.
Collapse
Affiliation(s)
- Mohamad Krayem
- Laboratory of Oncology and Experimental Surgery, Institut Jules Bordet, Université Libre de Bruxelles, 1000 Brussels, Belgium; (A.N.); (M.S.); (A.A.); (F.J.); (G.E.G.)
| | - Philippe Aftimos
- Medical Oncology Clinic, Institut Jules Bordet, Université Libre de Bruxelles, 1000 Brussels, Belgium; (P.A.); (J.K.)
| | - Ahmad Najem
- Laboratory of Oncology and Experimental Surgery, Institut Jules Bordet, Université Libre de Bruxelles, 1000 Brussels, Belgium; (A.N.); (M.S.); (A.A.); (F.J.); (G.E.G.)
| | - Tim van den Hooven
- PamGene International BV, 5211HH ’s-Hertogenbosch, The Netherlands; (T.v.d.H.); (A.v.d.B.); (L.H.-B.); (R.d.W.); (R.H.); (R.R.)
| | - Adriënne van den Berg
- PamGene International BV, 5211HH ’s-Hertogenbosch, The Netherlands; (T.v.d.H.); (A.v.d.B.); (L.H.-B.); (R.d.W.); (R.H.); (R.R.)
| | - Liesbeth Hovestad-Bijl
- PamGene International BV, 5211HH ’s-Hertogenbosch, The Netherlands; (T.v.d.H.); (A.v.d.B.); (L.H.-B.); (R.d.W.); (R.H.); (R.R.)
| | - Rik de Wijn
- PamGene International BV, 5211HH ’s-Hertogenbosch, The Netherlands; (T.v.d.H.); (A.v.d.B.); (L.H.-B.); (R.d.W.); (R.H.); (R.R.)
| | - Riet Hilhorst
- PamGene International BV, 5211HH ’s-Hertogenbosch, The Netherlands; (T.v.d.H.); (A.v.d.B.); (L.H.-B.); (R.d.W.); (R.H.); (R.R.)
| | - Rob Ruijtenbeek
- PamGene International BV, 5211HH ’s-Hertogenbosch, The Netherlands; (T.v.d.H.); (A.v.d.B.); (L.H.-B.); (R.d.W.); (R.H.); (R.R.)
| | - Malak Sabbah
- Laboratory of Oncology and Experimental Surgery, Institut Jules Bordet, Université Libre de Bruxelles, 1000 Brussels, Belgium; (A.N.); (M.S.); (A.A.); (F.J.); (G.E.G.)
| | - Joseph Kerger
- Medical Oncology Clinic, Institut Jules Bordet, Université Libre de Bruxelles, 1000 Brussels, Belgium; (P.A.); (J.K.)
| | - Ahmad Awada
- Laboratory of Oncology and Experimental Surgery, Institut Jules Bordet, Université Libre de Bruxelles, 1000 Brussels, Belgium; (A.N.); (M.S.); (A.A.); (F.J.); (G.E.G.)
- Medical Oncology Clinic, Institut Jules Bordet, Université Libre de Bruxelles, 1000 Brussels, Belgium; (P.A.); (J.K.)
| | - Fabrice Journe
- Laboratory of Oncology and Experimental Surgery, Institut Jules Bordet, Université Libre de Bruxelles, 1000 Brussels, Belgium; (A.N.); (M.S.); (A.A.); (F.J.); (G.E.G.)
| | - Ghanem E. Ghanem
- Laboratory of Oncology and Experimental Surgery, Institut Jules Bordet, Université Libre de Bruxelles, 1000 Brussels, Belgium; (A.N.); (M.S.); (A.A.); (F.J.); (G.E.G.)
| |
Collapse
|
722
|
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
|
723
|
Monti M, Consoli F, Vescovi R, Bugatti M, Vermi W. Human Plasmacytoid Dendritic Cells and Cutaneous Melanoma. Cells 2020; 9:E417. [PMID: 32054102 PMCID: PMC7072514 DOI: 10.3390/cells9020417] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 02/05/2020] [Accepted: 02/07/2020] [Indexed: 12/12/2022] Open
Abstract
The prognosis of metastatic melanoma (MM) patients has remained poor for a long time. However, the recent introduction of effective target therapies (BRAF and MEK inhibitors for BRAFV600-mutated MM) and immunotherapies (anti-CTLA-4 and anti-PD-1) has significantly improved the survival of MM patients. Notably, all these responses are highly dependent on the fitness of the host immune system, including the innate compartment. Among immune cells involved in cancer immunity, properly activated plasmacytoid dendritic cells (pDCs) exert an important role, bridging the innate and adaptive immune responses and directly eliminating cancer cells. A distinctive feature of pDCs is the production of high amount of type I Interferon (I-IFN), through the Toll-like receptor (TLR) 7 and 9 signaling pathway activation. However, published data indicate that melanoma-associated escape mechanisms are in place to hijack pDC functions. We have recently reported that pDC recruitment is recurrent in the early phases of melanoma, but the entire pDC compartment collapses over melanoma progression. Here, we summarize recent advances on pDC biology and function within the context of melanoma immunity.
Collapse
Affiliation(s)
- Matilde Monti
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (M.M.); (R.V.); (M.B.)
| | - Francesca Consoli
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, Medical Oncology, University of Brescia at ASST-Spedali Civili, 25123 Brescia, Italy;
| | - Raffaella Vescovi
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (M.M.); (R.V.); (M.B.)
| | - Mattia Bugatti
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (M.M.); (R.V.); (M.B.)
| | - William Vermi
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (M.M.); (R.V.); (M.B.)
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
724
|
Hanks JE, Kovatch KJ, Ali SA, Roberts E, Durham AB, Smith JD, Bradford CR, Malloy KM, Boonstra PS, Lao CD, McLean SA. Sentinel Lymph Node Biopsy in Head and Neck Melanoma: Long-term Outcomes, Prognostic Value, Accuracy, and Safety. Otolaryngol Head Neck Surg 2020; 162:520-529. [PMID: 32041486 DOI: 10.1177/0194599819899934] [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] [Indexed: 02/06/2023]
Abstract
OBJECTIVE To evaluate the long-term outcomes of sentinel lymph node biopsy (SLNB) for head and neck cutaneous melanoma (HNCM). STUDY DESIGN Retrospective cohort study. SETTING Tertiary academic medical center. SUBJECTS AND METHODS Longitudinal review of a 356-patient cohort with HNCM undergoing SLNB from 1997 to 2007. RESULTS Descriptive characteristics included the following: age, 53.5 ± 19 years (mean ± SD); sex, 26.8% female; median follow-up, 4.9 years; and Breslow depth, 2.52 ± 1.87 mm. Overall, 75 (21.1%) patients had a positive SLNB. Among patients undergoing completion lymph node dissection following positive SLNB, 20 (27.4%) had at least 1 additional positive nonsentinel lymph node. Eighteen patients with local control and negative SLNB developed regional disease, indicating a false omission rate of 6.4%, including 10 recurrences in previously unsampled basins. Ten-year overall survival (OS) and melanoma-specific survival (MSS) were significantly greater in the negative sentinel lymph node (SLN) cohort (OS, 61% [95% CI, 0.549-0.677]; MSS, 81.9% [95% CI, 0.769-0.873]) than the positive SLN cohort (OS, 31% [95% CI, 0.162-0.677]; MSS, 60.3% [95% CI, 0.464-0.785]) and positive SLN/positive nonsentinel lymph node cohort (OS, 8.4% [95% CI, 0.015-0.474]; MSS, 9.6% [95% CI, 0.017-0.536]). OS was significantly associated with SLN positivity (hazard ratio [HR], 2.39; P < .01), immunosuppression (HR, 2.37; P < .01), angiolymphatic invasion (HR, 1.91; P < .01), and ulceration (HR, 1.86; P < .01). SLN positivity (HR, 3.13; P < .01), angiolymphatic invasion (HR, 3.19; P < .01), and number of mitoses (P = .0002) were significantly associated with MSS. Immunosuppression (HR, 3.01; P < .01) and SLN status (HR, 2.84; P < .01) were associated with recurrence-free survival, and immunosuppression was the only factor significantly associated with regional recurrence (HR, 6.59; P < .01). CONCLUSIONS Long-term follow up indicates that SLNB showcases durable accuracy, safety, and prognostic importance for cutaneous HNCM.
Collapse
Affiliation(s)
- John E Hanks
- Department of Otolaryngology-Head and Neck Surgery, Michigan Medicine, Ann Arbor, Michigan, USA
| | - Kevin J Kovatch
- Department of Otolaryngology-Head and Neck Surgery, Michigan Medicine, Ann Arbor, Michigan, USA
| | - S Ahmed Ali
- Department of Otolaryngology-Head and Neck Surgery, Michigan Medicine, Ann Arbor, Michigan, USA
| | - Emily Roberts
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan, USA
| | - Alison B Durham
- Department of Dermatology, Michigan Medicine, Ann Arbor, Michigan, USA
| | - Joshua D Smith
- Department of Otolaryngology-Head and Neck Surgery, Michigan Medicine, Ann Arbor, Michigan, USA
| | - Carol R Bradford
- Department of Otolaryngology-Head and Neck Surgery, Michigan Medicine, Ann Arbor, Michigan, USA.,University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Kelly M Malloy
- Department of Otolaryngology-Head and Neck Surgery, Michigan Medicine, Ann Arbor, Michigan, USA
| | - Philip S Boonstra
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan, USA
| | - Christopher D Lao
- Department of Medical Oncology, Michigan Medicine, Ann Arbor, Michigan, USA
| | - Scott A McLean
- Department of Otolaryngology-Head and Neck Surgery, Michigan Medicine, Ann Arbor, Michigan, USA
| |
Collapse
|
725
|
Abstract
OPINION STATEMENT Patients with clinical stage III melanoma, defined as palpable lymph nodes with or without in-transit metastases, have poor prognosis even with recent advances with targeted and checkpoint inhibitor therapy in the adjuvant setting. Neoadjuvant therapy for clinical stage III melanoma is an attractive treatment paradigm as patient outcomes may be improved by earlier introduction to systemic therapy. Additionally, preoperative therapy that shrinks disease has the potential to improve surgical morbidity. Neoadjuvant therapy also provides for pathologic response assessment which can serve as a way to stratify patient outcomes and subsequent disease relapse risk. Early trials of neoadjuvant immunotherapy are yielding promising results, with high rates of pathologic complete response (pCR) and improved relapse-free survival rates. Ipilimumab, nivolumab with or without ipilimumab, and pembrolizumab have been investigated in the neoadjuvant setting. A meta-analysis has shown a 1-year relapse-free survival rate of over 80% with neoadjuvant immunotherapy. Importantly, pooled data also shows that pCR strongly correlates with outcomes. Early phase trials have also highlighted the importance of dosing of neoadjuvant therapy to appropriately balance response and immune related toxicities, which can be severe. The combination of ipilimumab 1 mg/kg and nivolumab 3 mg/kg has been identified as an optimal regimen for further study. Translational studies have highlighted the ability of neoadjuvant immunotherapy to expand tumor-specific T cells in both the tumor microenvironment and peripheral blood. At this time, surgical resection and adjuvant therapy remains standard of care for clinical stage III melanoma; however, appropriate patients should be considered for ongoing neoadjuvant clinical trials.
Collapse
Affiliation(s)
- Meredith S Pelster
- Department of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Rodabe N Amaria
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd. #430, Houston, TX, 77030, USA.
| |
Collapse
|
726
|
Kubota N, Taniguchi F, Nyuya A, Umeda Y, Mori Y, Fujiwara T, Tanioka H, Tsuruta A, Yamaguchi Y, Nagasaka T. Upregulation of microRNA-31 is associated with poor prognosis in patients with advanced colorectal cancer. Oncol Lett 2020; 19:2685-2694. [PMID: 32218819 PMCID: PMC7068240 DOI: 10.3892/ol.2020.11365] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 11/14/2019] [Indexed: 01/01/2023] Open
Abstract
Colorectal cancer (CRC) manifests after the accumulation of genetic and epigenetic alterations along with tumor microenvironments. MicroRNA (miRNA/miR) molecules have been revealed to serve in critical roles in the progression various types of cancer, and their expression level is often an important diagnostic, predictive or prognostic biomarker. The aim of the present study was to evaluate the potential of miRNAs as prognostic biomarkers for patients with advanced CRC. miRNA arrays were performed on CRC specimens obtained from tumors with various molecular statuses [e.g. KRAS proto-oncogene, GTPase (KRAS)/B-Raf proto-oncogene, serine/threonine kinase (BRAF)/microsatellite instability (MSI)], and their paired normal mucosal specimens. The miRNA array revealed that miR-31-5p (miR-31) was specifically upregulated in CRCs with the BRAF V600E mutation, the results of which were supported by subsequent analysis of a dataset retrieved from The Cancer Genome Atlas (TCGA) database, which contained information regarding 170 patients with CRC including 51 BRAF-mutant CRCs. Of our cohort of 67 patients with stage IV CRC, 15 (22%) and 4 (6%) showed KRAS and BRAF V600E mutations, respectively. Since the median miR-31 expression was 3.45 (range, 0.004–6330.531), the cut-off value was chosen as 3.5, and all tumors were categorized into two groups accordingly (high-/low-miR-31 expression). The high miR-31 expression group (n=33) was significantly associated with a poorer mortality (univariate hazard ratio=2.12; 95% confidence interval, 0.23–0.95; P=0.03) and exhibited a shorter median survival time (MST; 20.1 months) compared with the low miR-31 expression group (n=34) (MST, 38.3 months; P=0.03), indicating that miR-31 is a promising prognostic biomarker for patients with advanced CRC. Thus, performing a functional analysis of miR-31 expression may lead to the development of new targeted therapies for the various genetic subtypes of CRC.
Collapse
Affiliation(s)
- Nobuhito Kubota
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Fumitaka Taniguchi
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Akihiro Nyuya
- Department of Clinical Oncology, Kawasaki Medical School, Kurashiki, Okayama 701-0192, Japan
| | - Yuzo Umeda
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Yoshiko Mori
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Toshiyoshi Fujiwara
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Hiroaki Tanioka
- Department of Clinical Oncology, Kawasaki Medical School, Kurashiki, Okayama 701-0192, Japan
| | - Atsushi Tsuruta
- Department of Digestive Surgery, Kawasaki Medical School, Kurashiki, Okayama 701-0192, Japan
| | - Yoshiyuki Yamaguchi
- Department of Clinical Oncology, Kawasaki Medical School, Kurashiki, Okayama 701-0192, Japan
| | - Takeshi Nagasaka
- Department of Clinical Oncology, Kawasaki Medical School, Kurashiki, Okayama 701-0192, Japan
| |
Collapse
|
727
|
Karakousis G. Adjuvant therapy for melanoma: how to choose? Lancet Oncol 2020; 21:319-320. [PMID: 32007137 DOI: 10.1016/s1470-2045(20)30002-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Accepted: 01/02/2020] [Indexed: 10/25/2022]
Affiliation(s)
- Giorgos Karakousis
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, PA 19130, USA.
| |
Collapse
|
728
|
Biomarkers Predictive of Survival and Response to Immune Checkpoint Inhibitors in Melanoma. Am J Clin Dermatol 2020; 21:1-11. [PMID: 31602560 DOI: 10.1007/s40257-019-00475-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Immunotherapy has revolutionized the treatment of melanoma. Targeting of the immune checkpoints cytotoxic T-lymphocyte-associated protein 4 and programmed cell death protein 1 has led to improved survival in a subset of patients. Unfortunately, the use of immune checkpoint inhibitors is associated with significant side effects and many patients do not respond to treatment. Thus, there is an urgent need both for prognostic biomarkers to estimate risk and for predictive biomarkers to determine which patients are likely to respond to therapy. In this review, prognostic and predictive biomarkers that are an active area of research are outlined. Of note, certain transcriptomic signatures are already used in the clinic, albeit not routinely, to prognosticate patients. In the predictive setting, programmed cell death protein ligand 1 expression has been shown to correlate with benefit but is not precise enough to be used as an exclusionary biomarker. Future investigation will need to focus on biomarkers that are easily reproducible, cost effective, and accurate. The use of readily available clinical material, such as serum or hematoxylin and eosin-stained images, may offer one such path forward.
Collapse
|
729
|
Ghilu S, Li Q, Fontaine SD, Santi DV, Kurmasheva RT, Zheng S, Houghton PJ. Prospective use of the single-mouse experimental design for the evaluation of PLX038A. Cancer Chemother Pharmacol 2020; 85:251-263. [PMID: 31927611 PMCID: PMC7039322 DOI: 10.1007/s00280-019-04017-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 12/17/2019] [Indexed: 12/12/2022]
Abstract
PURPOSE Defining robust criteria for drug activity in preclinical studies allows for fewer animals per treatment group, and potentially allows for inclusion of additional cancer models that more accurately represent genetic diversity and, potentially, allows for tumor sensitivity biomarker identification. METHODS Using a single-mouse design, 32 pediatric xenograft tumor models representing diverse pediatric cancer types [Ewing sarcoma (9), brain (4), rhabdomyosarcoma (10), Wilms tumor (4), and non-CNS rhabdoid tumors (5)] were evaluated for response to a single administration of pegylated-SN38 (PLX038A), a controlled-release PEGylated formulation of SN-38. Endpoints measured were percent tumor regression, and event-free survival (EFS). The correlation between response to PLX038A was compared to that for ten models treated with irinotecan (2.5 mg/kg × 5 days × 2 cycles), using a traditional design (10 mice/group). Correlations between tumor sensitivity, genetic mutations and gene expression were sought. Models showing no disease at week 20 were categorized as 'extreme responders' to PLX038A, whereas those with EFS less than 5 weeks were categorized as 'resistant'. RESULTS The activity of PLX038A was evaluable in 31/32 models. PLX038A induced > 50% volume regressions in 25 models (78%). Initial tumor volume regression correlated only modestly with EFS (r2 = 0.238), but sensitivity to PLX038A was better correlated with response to irinotecan when one tumor hypersensitive to PLX038A was omitted (r2 = 0.6844). Mutations in 53BP1 were observed in three of six sensitive tumor models compared to none in resistant models (n = 6). CONCLUSIONS This study demonstrates the feasibility of using a single-mouse design for assessing the antitumor activity of an agent, while encompassing greater genetic diversity representative of childhood cancers. PLX038A was highly active in most xenograft models, and tumor sensitivity to PLX038A was correlated with sensitivity to irinotecan, validating the single-mouse design in identifying agents with the same mechanism of action. Biomarkers that correlated with model sensitivity included wild-type TP53, or mutant TP53 but with a mutation in 53BP1, thus a defect in DNA damage response. These results support the value of the single-mouse experimental design.
Collapse
Affiliation(s)
- Samson Ghilu
- Greehey Children's Cancer Research Institute, UT Health San Antonio, 8403 Floyd Curl Drive, San Antonio, TX, 78229, USA
| | - Qilin Li
- Greehey Children's Cancer Research Institute, UT Health San Antonio, 8403 Floyd Curl Drive, San Antonio, TX, 78229, USA
| | - Shaun D Fontaine
- ProLynx LLC, 455 Mission Bay Blvd, South San Francisco, CA, 94158, USA
| | - Daniel V Santi
- ProLynx LLC, 455 Mission Bay Blvd, South San Francisco, CA, 94158, USA
| | - Raushan T Kurmasheva
- Greehey Children's Cancer Research Institute, UT Health San Antonio, 8403 Floyd Curl Drive, San Antonio, TX, 78229, USA
| | - Siyuan Zheng
- Greehey Children's Cancer Research Institute, UT Health San Antonio, 8403 Floyd Curl Drive, San Antonio, TX, 78229, USA
| | - Peter J Houghton
- Greehey Children's Cancer Research Institute, UT Health San Antonio, 8403 Floyd Curl Drive, San Antonio, TX, 78229, USA.
| |
Collapse
|
730
|
Sullivan RJ, Hollebecque A, Flaherty KT, Shapiro GI, Rodon Ahnert J, Millward MJ, Zhang W, Gao L, Sykes A, Willard MD, Yu D, Schade AE, Crowe K, Flynn DL, Kaufman MD, Henry JR, Peng SB, Benhadji KA, Conti I, Gordon MS, Tiu RV, Hong DS. A Phase I Study of LY3009120, a Pan-RAF Inhibitor, in Patients with Advanced or Metastatic Cancer. Mol Cancer Ther 2020; 19:460-467. [PMID: 31645440 DOI: 10.1158/1535-7163.mct-19-0681] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 08/23/2019] [Accepted: 10/18/2019] [Indexed: 02/03/2023]
Abstract
Mutations in ERK signaling drive a significant percentage of malignancies. LY3009120, a pan-RAF and dimer inhibitor, has preclinical activity in RAS- and BRAF-mutated cell lines including BRAF-mutant melanoma resistant to BRAF inhibitors. This multicenter, open-label, phase I clinical trial (NCT02014116) consisted of part A (dose escalation) and part B (dose confirmation) in patients with advanced/metastatic cancer. In part A, oral LY3009120 was dose escalated from 50 to 700 mg twice a day on a 28-day cycle. In part B, 300 mg LY3009120 was given twice a day. The primary objective was to identify a recommended phase II dose (RP2D). Secondary objectives were to evaluate safety, pharmacokinetics, and preliminary efficacy. Identification of pharmacodynamic biomarkers was exploratory. In parts A and B, 35 and 16 patients were treated, respectively (N = 51). In part A, 6 patients experienced eight dose-limiting toxicities. The RP2D was 300 mg twice a day. Common (>10%) any-grade drug-related treatment-emergent adverse events were fatigue (n = 15), nausea (n = 12), dermatitis acneiform (n = 10), decreased appetite (n = 7), and maculopapular rash (n = 7). The median duration of treatment was 4 weeks; 84% of patients completed one or two cycles of treatment. Exposures observed at 300 mg twice a day were above the preclinical concentration associated with tumor regression. Eight patients had a best overall response of stable disease; there were no complete or partial clinical responses. Despite adequate plasma exposure levels, predicted pharmacodynamic effects were not observed.
Collapse
Affiliation(s)
- Ryan J Sullivan
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts
| | - Antoine Hollebecque
- Drug Development Department (DITEP), Gustave Roussy Cancer Institute, Villejuif, France
| | - Keith T Flaherty
- Developmental Therapeutics, Massachusetts General Hospital Cancer Center, Boston, Massachusetts
| | | | | | - Michael J Millward
- Linear Clinical Research, University of Western Australia, Perth, Australia
| | - Wei Zhang
- Eli Lilly and Company, Indianapolis, Indiana
| | - Ling Gao
- Eli Lilly and Company, Branchburg, New Jersey
| | | | | | - Danni Yu
- Eli Lilly and Company, Indianapolis, Indiana
| | | | | | | | | | | | | | | | | | | | - Ramon V Tiu
- Eli Lilly and Company, Indianapolis, Indiana
| | - David S Hong
- Department of Investigational Cancer Therapeutics, University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
731
|
Petrella TM, Fletcher GG, Knight G, McWhirter E, Rajagopal S, Song X, Baetz TD. Systemic adjuvant therapy for adult patients at high risk for recurrent cutaneous or mucosal melanoma: an Ontario Health (Cancer Care Ontario) clinical practice guideline. Curr Oncol 2020; 27:e43-e52. [PMID: 32218667 PMCID: PMC7096195 DOI: 10.3747/co.27.5933] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Background Previous versions of the guideline from the Program in Evidence-Based Care (pebc) at Ontario Health (Cancer Care Ontario) recommended that the use of high-dose interferon alfa 2b therapy be discussed and offered to patients with resected cutaneous melanoma with a high risk of recurrence. Subsequently, several clinical trials in patients with resected or metastatic melanoma found that immune checkpoint inhibitors and targeted therapies have a benefit greater than that with interferon. It was therefore considered timely for an update to the guideline about adjuvant systemic therapy in melanoma. Methods The present guideline was developed by the pebc and the Melanoma Disease Site Group (dsg). Based on a systematic review from a literature search conducted using medline, embase, and the Evidence Based Medicine Reviews databases for the period 1996 to 28 May 2019, the Working Group drafted recommendations. The systematic review and recommendations were then circulated to the Melanoma dsg and the pebc Report Approval Panel for internal review; the revised document underwent external review. Recommendations For patients with completely resected cutaneous or mucosal melanoma with a high risk of recurrence, the recommended adjuvant therapies are nivolumab, pembrolizumab, or dabrafenib-trametinib for patients with BRAF V600E or V600K mutations; nivolumab or pembrolizumab are recommend for patients with BRAF wild-type disease. Use of ipilimumab is not recommended. Molecular testing should be conducted to help guide treatment decisions. Interferon alfa, chemotherapy regimens, vaccines, levamisole, bevacizumab, bacillus Calmette-Guérin, and isolated limb perfusion are not recommended for adjuvant treatment of cutaneous melanoma except as part of a clinical trial.
Collapse
Affiliation(s)
- T M Petrella
- University of Toronto and Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, ON
| | - G G Fletcher
- Program in Evidence-Based Care, Ontario Health (Cancer Care Ontario), and Department of Oncology, McMaster University, Hamilton, ON
| | - G Knight
- Department of Oncology, McMaster University, Hamilton, and Grand River Regional Cancer Centre, Kitchener, ON
| | - E McWhirter
- Department of Oncology, Division of Medical Oncology, McMaster University, and Juravinski Cancer Centre, Hamilton, ON
| | | | - X Song
- Department of Internal Medicine, Division of Medical Oncology, University of Ottawa, and The Ottawa Hospital Cancer Centre, Ottawa, ON
| | - T D Baetz
- Department of Oncology, Queen's University, and Cancer Centre of Southeastern Ontario-Kingston General Hospital, Kingston, ON
| |
Collapse
|
732
|
Nakamura Y, Asai J, Igaki H, Inozume T, Namikawa K, Hayashi A, Fukushima S, Fujimura T, Ito T, Imafuku K, Tanaka R, Teramoto Y, Minagawa A, Miyagawa T, Miyashita A, Wada M, Koga H, Sugaya M. Japanese Dermatological Association Guidelines: Outlines of guidelines for cutaneous melanoma 2019. J Dermatol 2020; 47:89-103. [PMID: 31782186 DOI: 10.1111/1346-8138.15151] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 10/18/2019] [Indexed: 11/30/2022]
Abstract
With consideration of the ongoing developments in treatment options for cutaneous melanoma, the Japanese Skin Cancer Society published the first guidelines for cutaneous melanoma in 2007 and later revised them in 2015. Here, we report on an English version of the 2019 Japanese Melanoma Guidelines. In this latest edition, all processes were carried out according to the Grading of Recommendations, Assessment, Development and Evaluation system. A comprehensive published work search, systematic review and determination of recommendations in each clinical question were performed by a multidisciplinary expert panel consisting of dermatologists, a plastic and reconstructive surgeon, and a radiation oncologist. The advent of novel agents, such as immune checkpoint inhibitors and molecular-targeted agents, has drastically changed the nature of treatment for adjuvant and advanced-stage diseases among melanoma patients worldwide. Additionally, recent reports of clinical trials regarding surgical procedures and a better understanding of molecular biology and tumor immunology in clinical types of melanoma have had an impact on clinical practise. Based on these viewpoints, eight relevant clinical questions were raised in this report that aim to help clinicians select the appropriate therapeutic approach.
Collapse
Affiliation(s)
- Yasuhiro Nakamura
- Department of Skin Oncology/Dermatology, Saitama Medical University International Medical Center, Saitama, Japan
| | - Jun Asai
- Department of Dermatology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hiroshi Igaki
- Department of Radiation Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Takashi Inozume
- Department of Dermatology, University of Yamanashi, Kofu, Japan
| | - Kenjiro Namikawa
- Department of Dermatologic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Ayato Hayashi
- Department of Plastic and Reconstructive Surgery, Juntendo University Urayasu Hospital, Urayasu, Japan
| | - Satoshi Fukushima
- Department of Dermatology/Plastic and Reconstructive Surgery, Kumamoto University, Kumamoto, Japan
| | - Taku Fujimura
- Department of Dermatology, Tohoku University, Sendai, Japan
| | - Takamichi Ito
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Keisuke Imafuku
- Department of Dermatology, Hokkaido University, Sapporo, Japan
| | - Ryota Tanaka
- Deparmtent of Dermatology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Yukiko Teramoto
- Department of Skin Oncology/Dermatology, Saitama Medical University International Medical Center, Saitama, Japan
| | - Akane Minagawa
- Department of Dermatology, Shinshu University, Matsumoto, Japan
| | - Takuya Miyagawa
- Department of Dermatology, University of Tokyo, Tokyo, Japan
| | - Azusa Miyashita
- Department of Dermatology/Plastic and Reconstructive Surgery, Kumamoto University, Kumamoto, Japan
| | - Makoto Wada
- Department of Dermatology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hiroshi Koga
- Department of Dermatology, Shinshu University, Matsumoto, Japan
| | - Makoto Sugaya
- Department of Dermatology, International University of Health and Welfare, Narita, Japan
| |
Collapse
|
733
|
Erkes DA, Cai W, Sanchez IM, Purwin TJ, Rogers C, Field CO, Berger AC, Hartsough EJ, Rodeck U, Alnemri ES, Aplin AE. Mutant BRAF and MEK Inhibitors Regulate the Tumor Immune Microenvironment via Pyroptosis. Cancer Discov 2020; 10:254-269. [PMID: 31796433 PMCID: PMC7007378 DOI: 10.1158/2159-8290.cd-19-0672] [Citation(s) in RCA: 322] [Impact Index Per Article: 64.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 10/23/2019] [Accepted: 11/26/2019] [Indexed: 11/16/2022]
Abstract
Combinations of BRAF inhibitors and MEK inhibitors (BRAFi + MEKi) are FDA-approved to treat BRAF V600E/K-mutant melanoma. Efficacy of BRAFi + MEKi associates with cancer cell death and alterations in the tumor immune microenvironment; however, the links are poorly understood. We show that BRAFi + MEKi caused durable melanoma regression in an immune-mediated manner. BRAFi + MEKi treatment promoted cleavage of gasdermin E (GSDME) and release of HMGB1, markers of pyroptotic cell death. GSDME-deficient melanoma showed defective HMGB1 release, reduced tumor-associated T cell and activated dendritic cell infiltrates in response to BRAFi + MEKi, and more frequent tumor regrowth after drug removal. Importantly, BRAFi + MEKi-resistant disease lacked pyroptosis markers and showed decreased intratumoral T-cell infiltration but was sensitive to pyroptosis-inducing chemotherapy. These data implicate BRAFi + MEKi-induced pyroptosis in antitumor immune responses and highlight new therapeutic strategies for resistant melanoma. SIGNIFICANCE: Targeted inhibitors and immune checkpoint agents have advanced the care of patients with melanoma; however, detailed knowledge of the intersection between these two research areas is lacking. We describe a molecular mechanism of targeted inhibitor regulation of an immune-stimulatory form of cell death and provide a proof-of-principle salvage therapy concept for inhibitor-resistant melanoma.See related commentary by Smalley, p. 176.This article is highlighted in the In This Issue feature, p. 161.
Collapse
Affiliation(s)
- Dan A Erkes
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Weijia Cai
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Ileine M Sanchez
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Timothy J Purwin
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Corey Rogers
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Conroy O Field
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Adam C Berger
- Department of Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Edward J Hartsough
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania
| | - Ulrich Rodeck
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
- Department of Dermatology and Cutaneous Biology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Emad S Alnemri
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, Pennsylvania.
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Andrew E Aplin
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania.
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| |
Collapse
|
734
|
Adjuvant dabrafenib plus trametinib versus placebo in patients with resected, BRAF V600-mutant, stage III melanoma (COMBI-AD): exploratory biomarker analyses from a randomised, phase 3 trial. Lancet Oncol 2020; 21:358-372. [PMID: 32007138 DOI: 10.1016/s1470-2045(20)30062-0] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 11/29/2019] [Accepted: 12/02/2019] [Indexed: 01/01/2023]
Abstract
BACKGROUND Adjuvant dabrafenib plus trametinib reduced the risk of relapse versus placebo in patients with resected, BRAFV600-mutant, stage III melanoma in the phase 3 COMBI-AD trial. This prespecified exploratory biomarker analysis aimed to evaluate potential prognostic or predictive factors and mechanisms of resistance to adjuvant targeted therapy. METHODS COMBI-AD is a randomised, double-blind, placebo-controlled, phase 3 trial comparing dabrafenib 150 mg orally twice daily plus trametinib 2 mg orally once daily versus two matched placebos. Study participants were at least 18 years of age and underwent complete resection of stage IIIA (lymph node metastases >1 mm), IIIB, or IIIC cutaneous melanoma, per American Joint Committee on Cancer 7th edition criteria, with a BRAFV600E or BRAFV600K mutation. Patients were randomly assigned (1:1) to the two treatment groups by an interactive voice response system, stratified by mutation type and disease stage. Patients, physicians, and the investigators who analysed data were masked to treatment allocation. The primary outcome was relapse-free survival, defined as the time from randomisation to disease recurrence or death from any cause. Biomarker assessment was a prespecified exploratory outcome of the trial. We assessed intrinsic tumour genomic features by use of next-generation DNA sequencing and characteristics of the tumour microenvironment by use of a NanoString RNA assay, which might provide prognostic and predictive information. This trial is registered with ClinicalTrials.gov, number NCT01682083, and is ongoing but no longer recruiting participants. FINDINGS Between Jan 31, 2013, and Dec 11, 2014, 870 patients were enrolled in the trial. Median follow-up at data cutoff (April 30, 2018) was 44 months (IQR 38-49) in the dabrafenib plus trametinib group and 42 months (21-49) in the placebo group. Intrinsic tumour genomic features were assessed in 368 patients (DNA sequencing set) and tumour microenvironment characteristics were assessed in 507 patients (NanoString biomarker set). MAPK pathway genomic alterations at baseline did not affect treatment benefit or clinical outcome. An IFNγ gene expression signature higher than the median was prognostic for prolonged relapse-free survival in both treatment groups. Tumour mutational burden was independently prognostic for relapse-free survival in the placebo group (high TMB, top third; hazard ratio [HR] 0·56, 95% CI 0·37-0·85, p=0·0056), but not in the dabrafenib plus trametinib group (0·83, 95% CI 0·53-1·32, p=0·44). Patients with tumour mutational burden in the lower two terciles seem to derive a substantial long-term relapse-free survival benefit from targeted therapy (HR [versus placebo] 0·49, 95% CI 0·35-0·68, p<0·0001). However, patients with high tumour mutational burden seem to have a less pronounced benefit with targeted therapy (HR [versus placebo] 0·75, 95% CI 0·44-1·26, p=0·27), especially if they had an IFNγ signature lower than the median (HR 0·88 [95% CI 0·40-1·93], p=0·74). INTERPRETATION Tumour mutational burden alone or in combination with IFNγ gene expression signature or other markers for an adaptive immune response might be of relevance for identifying patients with stage III melanoma who might derive clinical benefit from targeted therapy. Further validation in prospective clinical trials is warranted. FUNDING Novartis Pharmaceuticals.
Collapse
|
735
|
Echegaray JJ, Yeaney G, Chen R, Singh AD. Conjunctival Melanoma Angiotropic Microsatellitosis: A Mechanism of Local Extravascular Migratory Metastasis. Ocul Oncol Pathol 2020; 6:287-292. [PMID: 33005619 DOI: 10.1159/000505270] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 12/06/2019] [Indexed: 11/19/2022] Open
Abstract
Purpose To report a case of local metastasis of conjunctival melanoma, which may occur via extravascular migratory metastasis (EVMM), and discuss its clinical relevance in conjunctival melanoma tumor staging and possible management implications. Methods Retrospective chart review of a single clinical case with clinicopathologic correlation. Results A 65-year-old male referred due to local recurrence of conjunctival melanoma at the caruncle was successfully treated after two excisional procedures with negative sentinel lymph node biopsies. Forty-eight months after initial presentation, the patient developed a nodular lesion representing local recurrence in the ipsilateral upper tarsal conjunctiva, distant from the primary tumor site. Histopathology showed nodules in the substantia propria in the absence of primary acquired melanosis. The tumor cells were found along the extravascular surface without intralymphatic or intravascular tumor cells consistent with local metastasis. One possible mechanism is angiotropic microsatellitosis leading to local EVMM. Additional neck CT imaging showed no lymphadenopathy. Conclusion EVMM via angiotropic microsatellitosis is another possible mechanism of noncontiguous local recurrence of conjunctival melanoma. Angiotropic microsatellitosis may represent a high-risk finding possibly related to increased melanoma-related mortality.
Collapse
Affiliation(s)
- Jose J Echegaray
- Department of Ophthalmic Oncology, Cole Eye Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | | | - Rachel Chen
- Department of Ophthalmic Oncology, Cole Eye Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Arun D Singh
- Department of Ophthalmic Oncology, Cole Eye Institute, Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
736
|
Ziogas DC, Konstantinou F, Bouros S, Gogas H. Identifying the optimum first-line therapy in BRAF-mutant metastatic melanoma. Expert Rev Anticancer Ther 2020; 20:53-62. [PMID: 31903803 DOI: 10.1080/14737140.2020.1711737] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Introduction: The emergence of molecularly targeted agents and immune checkpoint inhibitors has positively revolutionized the management and prognosis of BRAF-mutant metastatic melanoma. However, the availability of both therapeutic options, with their pros and cons, rationally triggered clinical considerations for the optimum frontline and subsequent treatment decisions.Areas covered: Here, we debate all approved therapies in patients with BRAF-mutant metastatic melanoma evaluating their efficacy and safety based on their pivotal trials. With prospective randomized data pending, retrospective comparisons of BRAF/MEK versus immune checkpoint inhibitors are reviewed to recognize any advantage between these two alternatives and to optimize their implementation. Preclinical and early clinical results of combining concurrently or sequentially targeted therapy and immunotherapy are also discussed.Expert opinion: BRAF/MEK inhibitors produce rapid and deep responses and should be included in first-line approaches, particularly in cases with aggressive and bulky disease, while single or double checkpoint inhibition lead to more durable responses and could be involved either in frontline treatment of BRAF-mutant melanoma with less unfavorable characteristics or in maintenance after initial targeted induction or in future immune/targeted regimens for high-risk groups. Data from ongoing trials directly comparing or combining these strategies are expected to update their role in a more individualized basis.
Collapse
Affiliation(s)
- Dimitrios C Ziogas
- First Department of Medicine, National and Kapodistrian University of Athens, School of Medicine, Laiko General Hospital, Athens, Greece
| | - Frosso Konstantinou
- First Department of Medicine, National and Kapodistrian University of Athens, School of Medicine, Laiko General Hospital, Athens, Greece
| | - Spyros Bouros
- First Department of Medicine, National and Kapodistrian University of Athens, School of Medicine, Laiko General Hospital, Athens, Greece
| | - Helen Gogas
- First Department of Medicine, National and Kapodistrian University of Athens, School of Medicine, Laiko General Hospital, Athens, Greece
| |
Collapse
|
737
|
Freeley M. Current postgraduate training programs and online courses in precision medicine. Expert Rev Mol Diagn 2020; 20:569-574. [PMID: 31875486 DOI: 10.1080/14737159.2020.1709826] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Michael Freeley
- School of Biotechnology (Office X225), Dublin City University , Glasnevin, Ireland
| |
Collapse
|
738
|
Aderhold K, Wilson M, Berger AC, Levi S, Bennett J. Precision Medicine in the Treatment of Melanoma. Surg Oncol Clin N Am 2020; 29:1-13. [DOI: 10.1016/j.soc.2019.08.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
739
|
Eroglu Z, Eatrides J, Naqvi SMH, Kim Y, Rich J, Babacan NA, Brohl AS, Markowitz J, Sarnaik A, Zager J, Khushalani NI, Sondak VK, Messina J. Neoadjuvant BRAF-targeted therapy in regionally advanced and oligometastatic melanoma. Pigment Cell Melanoma Res 2020; 33:86-95. [PMID: 31329344 PMCID: PMC6928428 DOI: 10.1111/pcmr.12813] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Revised: 07/02/2019] [Accepted: 07/04/2019] [Indexed: 12/11/2022]
Abstract
Current management of locoregional and oligometastatic melanoma is typically with surgery; however, some patients are unable to undergo resection due to location/size of their tumors and/or the anticipated morbidity of the surgery. While there are currently no established guidelines for neoadjuvant therapy in melanoma, neoadjuvant BRAF-targeted therapy may make resection more feasible. A retrospective analysis was conducted of 23 patients with BRAFV600-mutant, stage III/IV melanoma treated with BRAF-targeted therapy prior to surgery, with no adjuvant treatment. Surgical specimens, preoperative imaging, and clinical outcomes were evaluated. Results: Ten of 23 patients (44%) attained a pathologic complete response (pCR), with no correlation between RECIST response based on preoperative imaging and pathologic response. After a median of 43-month follow-up, only 1 patient (10%) with a pCR recurred, while 8 of 13 (62%) patients without a pCR recurred. Patients with a pCR had significantly improved relapse-free (RFS) and overall survival (OS) compared to patients with residual tumor. Neoadjuvant BRAF-targeted therapy is associated with a high pCR rate in patients with stage III-IV melanoma, which may correlate with improved RFS and OS.
Collapse
Affiliation(s)
- Zeynep Eroglu
- Department of Cutaneous Oncology, Moffitt Cancer Center,
Tampa, FL
| | | | | | - Youngchul Kim
- Department of Biostatistics and Bioinformatics, Moffitt
Cancer Center, Tampa, FL
| | - Jeani Rich
- Department of Cutaneous Oncology, Moffitt Cancer Center,
Tampa, FL
| | | | - Andrew S Brohl
- Department of Cutaneous Oncology, Moffitt Cancer Center,
Tampa, FL
| | - Joseph Markowitz
- Department of Cutaneous Oncology, Moffitt Cancer Center,
Tampa, FL
| | - Amod Sarnaik
- Department of Cutaneous Oncology, Moffitt Cancer Center,
Tampa, FL
| | - Jonathan Zager
- Department of Cutaneous Oncology, Moffitt Cancer Center,
Tampa, FL
| | | | - Vernon K Sondak
- Department of Cutaneous Oncology, Moffitt Cancer Center,
Tampa, FL
| | - Jane Messina
- Department of Cutaneous Oncology, Moffitt Cancer Center,
Tampa, FL
| |
Collapse
|
740
|
Amaral TMS, Hoffmann MC, Sinnberg T, Niessner H, Sülberg H, Eigentler TK, Garbe C. Clinical validation of a prognostic 11-gene expression profiling score in prospectively collected FFPE tissue of patients with AJCC v8 stage II cutaneous melanoma. Eur J Cancer 2020; 125:38-45. [PMID: 31838403 DOI: 10.1016/j.ejca.2019.10.027] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 10/18/2019] [Accepted: 10/28/2019] [Indexed: 01/08/2023]
Abstract
BACKGROUND Adjuvant therapies have been approved for patients with AJCC (American Joint Committee on Cancer) stage III and stage IV cutaneous melanoma (CM) after complete resection. These therapies might also be indicated for patients with high-risk stage II CM. MATERIAL AND METHODS We included patients diagnosed with stage II melanoma between 2000 and 2016 and for which primary tumour tissue was available. The prognostic value of the 11-gene expression profiling score (GEPS) was evaluated as a dichotomized parameter (GEPS ≤0 vs. >0). Endpoints of the analysis were melanoma specific survival (MSS), distant metastasis-free survival (DMFS) and relapse-free survival (RFS). RESULTS GEPS was determined in 245 patients ranging between -0.7 and 3.53. A total of 111 females and 134 males were included; the median follow-up was 41 months. Kaplan Meier analyses showed statistically significant survival differences between patients with high GEPS (n = 154) and low GEPS (n = 91) for MSS (p = 0.018), DMFS (p = 0.005) and RFS (p = 0.009). The 5-year and 10-year MSS was 92% in the low-GEPS and 82% and 67% in the high-GEPS group, respectively. Multivariate Cox regression analysis showed independent significance for MSS of GEPS (HR = 1.55; p = 0.006), tumor thickness (HR = 1.21; p < 0.001) and age (HR1.05; p = 0.002). CONCLUSION GEPS was validated as independent prognostic factor for MSS in stage II CM and could be used for therapeutic decisions when systemic therapies become available in stage II CM.
Collapse
Affiliation(s)
- Teresa M S Amaral
- Center for Dermatooncology, Department of Dermatology, Eberhard Karls University of Tuebingen, Tuebingen, Germany; Portuguese Air Force, Health Care Direction, Lisbon, Portugal
| | | | - Tobias Sinnberg
- Center for Dermatooncology, Department of Dermatology, Eberhard Karls University of Tuebingen, Tuebingen, Germany
| | - Heike Niessner
- Center for Dermatooncology, Department of Dermatology, Eberhard Karls University of Tuebingen, Tuebingen, Germany
| | - Heiko Sülberg
- X-act Cologne Clinical Research GmbH, Cologne, Germany
| | - Thomas K Eigentler
- Center for Dermatooncology, Department of Dermatology, Eberhard Karls University of Tuebingen, Tuebingen, Germany
| | - Claus Garbe
- Center for Dermatooncology, Department of Dermatology, Eberhard Karls University of Tuebingen, Tuebingen, Germany; Central Malignant Melanoma Registry (CMMR) of Germany, Tuebingen, Germany.
| |
Collapse
|
741
|
The challenge of identifying which stage III melanoma patients need adjuvant treatment and with what. Ann Oncol 2020; 31:11-12. [DOI: 10.1016/j.annonc.2019.10.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 10/25/2019] [Indexed: 11/22/2022] Open
|
742
|
Strub T, Ballotti R, Bertolotto C. The "ART" of Epigenetics in Melanoma: From histone "Alterations, to Resistance and Therapies". Theranostics 2020; 10:1777-1797. [PMID: 32042336 PMCID: PMC6993228 DOI: 10.7150/thno.36218] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 11/14/2019] [Indexed: 02/07/2023] Open
Abstract
Malignant melanoma is the most deadly form of skin cancer. It originates from melanocytic cells and can also arise at other body sites. Early diagnosis and appropriate medical care offer excellent prognosis with up to 5-year survival rate in more than 95% of all patients. However, long-term survival rate for metastatic melanoma patients remains at only 5%. Indeed, malignant melanoma is known for its notorious resistance to most current therapies and is characterized by both genetic and epigenetic alterations. In cutaneous melanoma (CM), genetic alterations have been implicated in drug resistance, yet the main cause of this resistance seems to be non-genetic in nature with a change in transcription programs within cell subpopulations. This change can adapt and escape targeted therapy and immunotherapy cytotoxic effects favoring relapse. Because they are reversible in nature, epigenetic changes are a growing focus in cancer research aiming to prevent or revert the drug resistance with current therapies. As such, the field of epigenetic therapeutics is among the most active area of preclinical and clinical research with effects of many classes of epigenetic drugs being investigated. Here, we review the multiplicity of epigenetic alterations, mainly histone alterations and chromatin remodeling in both cutaneous and uveal melanomas, opening opportunities for further research in the field and providing clues to specifically control these modifications. We also discuss how epigenetic dysregulations may be exploited to achieve clinical benefits for the patients, the limitations of these therapies, and recent data exploring this potential through combinatorial epigenetic and traditional therapeutic approaches.
Collapse
Affiliation(s)
- Thomas Strub
- Université Nice Côte d'Azur, Inserm, C3M, France
- Biology and pathologies of melanocytes, Equipe labellisée ARC 2019, C3M, team 1, France
| | - Robert Ballotti
- Université Nice Côte d'Azur, Inserm, C3M, France
- Biology and pathologies of melanocytes, Equipe labellisée ARC 2019, C3M, team 1, France
| | - Corine Bertolotto
- Université Nice Côte d'Azur, Inserm, C3M, France
- Biology and pathologies of melanocytes, Equipe labellisée ARC 2019, C3M, team 1, France
| |
Collapse
|
743
|
Kashyap L, Saha S, Srikanth A. Dabrafenib: A narrative drug review. CANCER RESEARCH, STATISTICS, AND TREATMENT 2020. [DOI: 10.4103/crst.crst_210_20] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
744
|
Abstract
The pathological diagnosis of melanoma can be challenging. The provision of an appropriate biopsy and pertinent history can assist in establishing an accurate diagnosis and reliable estimate of prognosis. In their reports, pathologists should document both the criteria on which the diagnosis was based as well as important prognostic parameters. For melanoma, such prognostic parameters include tumor thickness, ulceration, mitotic rate, lymphovascular invasion, neurotropism, and tumor-infiltrating lymphocytes. Disease staging is important for risk stratifying melanoma patients into prognostic groups and patient management recommendations are often stage based. The 8th edition American Joint Committee on Cancer (AJCC) Melanoma Staging System was implemented in 2018 and several important changes were made. Tumor thickness and ulceration remain the key T category criteria. T1b melanomas were redefined as either ulcerated melanomas <1.0 mm thick or nonulcerated melanomas 0.8-1.0 mm thick. Although mitotic rate was removed as a T category criterion in the 8th edition, it remains a very important prognostic factor and should continue to be documented in primary melanoma pathology reports. It was also recommended in the 8th edition that tumor thickness be recorded to the nearest 0.1 mm (rather than the nearest 0.01 mm). In the future, incorporation of additional prognostic parameters beyond those utilized in the current version of the staging system into (web based) prognostic models/clinical tools will likely facilitate more personalized prognostic estimates. Evaluation of molecular markers of prognosis is an active area of current research; however, additional data are needed before it would be appropriate to recommend use of such tests in routine clinical practice.
Collapse
|
745
|
Bergdorf KN, Lee LA, Weiss VL. BRAF molecular testing in cytopathology: Implications for diagnosis, prognosis, and targeted therapeutics. Cancer Cytopathol 2020; 128:9-11. [PMID: 31765065 PMCID: PMC10089280 DOI: 10.1002/cncy.22209] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
| | - Laura A. Lee
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center
| | - Vivian L. Weiss
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center
- Correspondence should be addressed to: Vivian Lee Weiss, MD, Ph.D., Vanderbilt University Medical Center, CC-2213 MCN, Nashville, TN 37232. Phone: 615-875-3002, Fax: 615-343-7023,
| |
Collapse
|
746
|
Ge J, Wang J, Wang H, Jiang X, Liao Q, Gong Q, Mo Y, Li X, Li G, Xiong W, Zhao J, Zeng Z. The BRAF V600E mutation is a predictor of the effect of radioiodine therapy in papillary thyroid cancer. J Cancer 2020; 11:932-939. [PMID: 31949496 PMCID: PMC6959026 DOI: 10.7150/jca.33105] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Accepted: 10/20/2019] [Indexed: 02/07/2023] Open
Abstract
Objective: To investigate the correlation between the BRAF V600E gene mutation and clinicopathological features and thyroid function after iodine-131 treatment in patients with papillary thyroid cancer (PTC). Methods: A total of 128 PTC patients who underwent iodine-131 treatment after a total thyroidectomy from February 2015 to November 2016 at Hunan Cancer Hospital, China, were recruited. There were 25 males and 103 females. The age range was 11 to 73 years old. The BRAF V600E mutation in tumor tissues was detected by amplification-restriction mutation system polymerase chain reaction (ARMS-PCR), and the serum levels of Tg, TSH, Tg-Ab, and Tpo-Ab were measured by chemiluminescence after iodine-131 treatment. The BRAF V600E mutation was shown to be associated with clinicopathological characteristics and thyroid function indicators after iodine-131 treatment. Results: BRAF V600E mutation was detected in 75 of the 128 patients (58.6%) and was observed more frequently in cases with elevated Tg levels (Tg>1.00) at 3, 6, 12, and 18 months after treatment compared with patients without any BRAF mutations (P<0.05). Patients with BRAF V600E mutation had significant lower level of Tg-Ab at 3 and 12 months after treatment with iodine-131 than patients without BRAF V600E mutation (P<0.05). Among the 75 BRAF V600E patients, no significant association was found between the levels of TSH and Tpo-Ab after iodine-131 treatment (P>0.05). The BRAF V600E mutation was closely associated with the high-risk and age of the patient (≥45 years old) (P<0.05), but there was no significant correlation with gender, clinical stage, and distant metastasis. Conclusion: The BRAF V600E mutation is closely related to serum Tg elevation after treatment with iodine-131 in papillary thyroid cancer. These findings suggest that this BRAF mutation may be a predictor of the efficacy of iodine-131 treatment for papillary thyroid cancer.
Collapse
Affiliation(s)
- Junshang Ge
- NHC Key Laboratory of Carcinogenesis (Central South University) and Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China.,Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, the Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jie Wang
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, China
| | - Hui Wang
- NHC Key Laboratory of Carcinogenesis (Central South University) and Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Xianjie Jiang
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, China
| | - Qianjin Liao
- NHC Key Laboratory of Carcinogenesis (Central South University) and Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Qian Gong
- NHC Key Laboratory of Carcinogenesis (Central South University) and Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yongzhen Mo
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, China
| | - Xiaoling Li
- NHC Key Laboratory of Carcinogenesis (Central South University) and Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China.,Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, the Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Guiyuan Li
- NHC Key Laboratory of Carcinogenesis (Central South University) and Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China.,Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, the Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wei Xiong
- NHC Key Laboratory of Carcinogenesis (Central South University) and Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China.,Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, the Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jin Zhao
- NHC Key Laboratory of Carcinogenesis (Central South University) and Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China.,Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, China
| | - Zhaoyang Zeng
- NHC Key Laboratory of Carcinogenesis (Central South University) and Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China.,Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, the Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
747
|
Peach H, Board R, Cook M, Corrie P, Ellis S, Geh J, King P, Laitung G, Larkin J, Marsden J, Middleton M, Moncrieff M, Nathan P, Powell B, Pritchard-Jones R, Rodwell S, Steven N, Lorigan P. Current role of sentinel lymph node biopsy in the management of cutaneous melanoma: A UK consensus statement. J Plast Reconstr Aesthet Surg 2020; 73:36-42. [DOI: 10.1016/j.bjps.2019.06.020] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Accepted: 06/09/2019] [Indexed: 10/26/2022]
|
748
|
Weber J, Glutsch V, Geissinger E, Haug L, Lock JF, Schneider F, Kneitz H, Goebeler M, Schilling B, Gesierich A. Neoadjuvant immunotherapy with combined ipilimumab and nivolumab in patients with melanoma with primary or in transit disease. Br J Dermatol 2019; 183:559-563. [PMID: 31773720 DOI: 10.1111/bjd.18739] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/23/2019] [Indexed: 02/04/2023]
Abstract
The introduction of new therapeutic agents has revolutionized the treatment of metastatic melanoma. The approval of adjuvant anti-programmed death-1 monotherapy with nivolumab or pembrolizumab, and dabrafenib plus trametinib has recently set a new landmark in the treatment of stage III melanoma. Now, clinical trials have shown that immune checkpoint blockade can be performed in a neoadjuvant setting, an approach established as a standard therapeutic approach for other tumour entities such as breast cancer. Recent studies suggest that a pathological response achieved by neoadjuvant immunotherapy is associated with long-term tumour control and that short neoadjuvant application of checkpoint inhibitors may be superior to adjuvant therapy. Most recently, neoadjuvant ipilimumab plus nivolumab in stage III melanoma was reported. With two courses of dose-optimized ipilimumab (1 mg kg-1 ) combined with nivolumab (3 mg kg-1 ), pathological responses were observed in 77% of patients, while only 20% of patients experienced grade 3 or 4 adverse events. However, the neoadjuvant trials employing combined immune checkpoint blockade conducted so far have excluded patients with in transit metastases, a common finding in stage III melanoma. Here we report four patients with in transit metastases or an advanced primary tumour who have been treated with neoadjuvant ipilimumab plus nivolumab according to the OpACIN-neo trial scheme (arm B). All patients achieved radiological disease control and a pathological response. None of the patients has relapsed so far. Linked Comment: Blankenstein and van Akkooi. Br J Dermatol 2020; 183:421-422.
Collapse
Affiliation(s)
- J Weber
- Department of Dermatology, Venereology and Allergology, University Hospital Würzburg, Würzburg, Germany
| | - V Glutsch
- Department of Dermatology, Venereology and Allergology, University Hospital Würzburg, Würzburg, Germany
| | - E Geissinger
- Institute of Pathology, Julius-Maximilians-University, Würzburg, Germany
| | - L Haug
- Institute of Pathology, Julius-Maximilians-University, Würzburg, Germany
| | - J F Lock
- Department of General, Visceral, Transplant, Vascular and Paediatric Surgery, University Hospital Würzburg, Würzburg, Germany
| | - F Schneider
- Institute for Diagnostic and Interventional Radiology, University Hospital Würzburg, Würzburg, Germany
| | - H Kneitz
- Department of Dermatology, Venereology and Allergology, University Hospital Würzburg, Würzburg, Germany
| | - M Goebeler
- Department of Dermatology, Venereology and Allergology, University Hospital Würzburg, Würzburg, Germany
| | - B Schilling
- Department of Dermatology, Venereology and Allergology, University Hospital Würzburg, Würzburg, Germany
| | - A Gesierich
- Department of Dermatology, Venereology and Allergology, University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
749
|
Luke JJ, Ascierto PA, Carlino MS, Gershenwald JE, Grob JJ, Hauschild A, Kirkwood JM, Long GV, Mohr P, Robert C, Ross M, Scolyer RA, Yoon CH, Poklepovic A, Rutkowski P, Anderson JR, Ahsan S, Ibrahim N, M Eggermont AM. KEYNOTE-716: Phase III study of adjuvant pembrolizumab versus placebo in resected high-risk stage II melanoma. Future Oncol 2019; 16:4429-4438. [PMID: 31870188 DOI: 10.2217/fon-2019-0666] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Patients with high-risk stage II melanoma are at significant risk for recurrence after surgical resection. Adjuvant treatment options to lower the risk for distant metastases are limited. Although adjuvant IFN-α2b is associated with improved relapse-free survival in patients with high-risk melanoma, toxicity and limited overall survival benefits limit its use. Adjuvant treatment with the PD-1 inhibitor pembrolizumab significantly improved recurrence-free survival, compared with placebo, in patients with resected stage III melanoma in the Phase III KEYNOTE-054 trial; efficacy in patients with stage II disease has not been established. This article describes the design and rationale of KEYNOTE-716 (NCT03553836), a two-part, randomized, placebo-controlled, multicenter Phase III study of adjuvant pembrolizumab in patients with surgically resected high-risk stage II melanoma. Clinical trial registry & ID: ClinicalTrials.gov, NCT03553836.
Collapse
Affiliation(s)
- Jason J Luke
- Cancer Immunotherapeutics Center, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15232, USA
| | - Paolo A Ascierto
- Melanoma, Cancer Immunotherapy and Development Therapeutics Unit, Istituto Nazionale Tumori IRCCS "Fondazione G. Pascale," Naples, Italy
| | - Matteo S Carlino
- Department of Medical Oncology, Crown Princess Mary Cancer Centre, Westmead Hospital, Westmead, NSW, Australia.,Blacktown Cancer and Haematology Centre, Blacktown Hospital, Blacktown, NSW, Australia.,Melanoma Institute Australia, Sydney, NSW, Australia.,Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Jeffrey E Gershenwald
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jean-Jacques Grob
- Department of Dermatology and Skin Cancers, Hôpital de la Timone, Aix-Marseille Université, Marseille, France
| | - Axel Hauschild
- Department of Dermatological Oncology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - John M Kirkwood
- Cancer Immunotherapeutics Center, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15232, USA
| | - Georgina V Long
- Melanoma Institute Australia, Sydney, NSW, Australia.,Sydney Medical School, The University of Sydney, Sydney, NSW, Australia.,Department of Medical Oncology, Mater Hospital, North Sydney, NSW, Australia.,Department of Medical Oncology, Royal North Shore Hospital, St Leonards, NSW, Australia
| | - Peter Mohr
- Department of Dermatology, Elbe Kliniken Buxtehude, Buxtehude, Germany
| | - Caroline Robert
- Dermatology Unit, Department of Oncology, Gustave Roussy Cancer Centre, Villejuif, France.,Department of Medicine, Université Paris-Sud, Orsay, France
| | - Merrick Ross
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Richard A Scolyer
- Melanoma Institute Australia, Sydney, NSW, Australia.,Sydney Medical School, The University of Sydney, Sydney, NSW, Australia.,Department of Medicine, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Charles H Yoon
- Department of Surgical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Andrew Poklepovic
- Department of Internal Medicine, VCU Massey Cancer Center, Richmond, VA 23298, USA
| | - Piotr Rutkowski
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie Institute - Oncology Center, Warsaw, Poland
| | - James R Anderson
- Department of Clinical Oncology, Merck & Co., Inc., Kenilworth, NJ 07033, USA
| | - Sama Ahsan
- Department of Clinical Oncology, Merck & Co., Inc., Kenilworth, NJ 07033, USA
| | - Nageatte Ibrahim
- Department of Clinical Oncology, Merck & Co., Inc., Kenilworth, NJ 07033, USA
| | - Alexander M M Eggermont
- Dermatology Unit, Department of Oncology, Gustave Roussy Cancer Centre, Villejuif, France.,Department of Medical Oncology, Gustave Roussy Cancer Institute & Université Paris-Saclay, Paris, France
| |
Collapse
|
750
|
Poklepovic AS, Luke JJ. Considering adjuvant therapy for stage II melanoma. Cancer 2019; 126:1166-1174. [PMID: 31869447 PMCID: PMC7065103 DOI: 10.1002/cncr.32585] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 07/09/2019] [Accepted: 08/18/2019] [Indexed: 12/21/2022]
Abstract
Melanoma is among the few cancers that demonstrate an increasing incidence over time. Simultaneously, this trend has been marked by an epidemiologic shift to earlier stage at diagnosis. Before 2011, treatment options were limited for patients with metastatic disease, and the median overall survival was less than 1 year. Since then, the field of melanoma therapeutics has undergone major changes. The use of anti–CTLA‐4 and anti‐PD1 immune checkpoint inhibitors and combination BRAF/MEK inhibitors for patients with BRAF V600 mutations has significantly extended survival and allowed some patients to remain in durable disease remission off therapy. It has now been confirmed that these classes of agents have a benefit for patients with stage III melanoma after surgical resection, and anti‐PD1 and BRAF/MEK inhibitors are standards of care in this setting. Some patients with stage II disease (lymph node‐negative; American Joint Committee on Cancer stage IIB and IIC) have worse melanoma‐specific survival relative to some patients with stage III disease. Given these results, expanding the population of patients who are considered for adjuvant therapy to include those with stage II melanoma has become a priority, and randomized phase 3 clinical trials are underway. Moving into the future, the validation of patient risk‐stratification and treatment‐benefit prediction models will be important to improve the number needed to treat and limit exposure to toxicity in the large population of patients with early stage melanoma. Adjuvant therapy has improved outcomes in patients with stage III melanoma and is being explored in those with stage II melanoma. Stage III data as well as risk‐stratification tools and clinical considerations for the lymph node‐negative population are reviewed.
Collapse
Affiliation(s)
- Andrew S Poklepovic
- Department of Medicine, Virginia Commonwealth University, Richmond, Virginia.,Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia
| | - Jason J Luke
- Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania.,Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| |
Collapse
|